首页 > 最新文献

Cell Communication and Signaling最新文献

英文 中文
Tumor-microenvironment-on-a-chip: the construction and application. 肿瘤微环境芯片:构建与应用。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-23 DOI: 10.1186/s12964-024-01884-4
Hanzheng Xu, Jiangtao Wen, Jiahua Yang, Shufen Zhou, Yijie Li, Ke Xu, Wei Li, Sen Li

Currently, despite the vast amounts of time and money invested in cancer treatment, cancer remains one of the primary threats to human life. The primary factor contributing to the low treatment efficacy is cancer heterogeneity. The unclear molecular mechanisms underlying tumorigenesis, coupled with the complexity of human physiology, and the inability of animal models to accurately replicate the human tumor microenvironment, pose significant hurdles in the development of novel cancer therapies. Tumor-microenvironment-on-chip (TMOC) represents a research platform that integrates three-dimensional cell culture with microfluidic systems, simulating the essential components and physiological traits of the in vivo tumor microenvironment. It offers a dynamic setting within the chip system to study tumor progression, potentially heralding a breakthrough in cancer research. In this review, we will summarize the current advancements in this platform, encompassing various types of TMOCs and their applications in different types of cancer. From our perspective, the TMOC platform necessitates enhanced integration with tissue engineering techniques and microphysiological environments before it can evolve into a more refined preclinical model for cancer research.

目前,尽管在癌症治疗方面投入了大量的时间和金钱,但癌症仍然是人类生命的主要威胁之一。导致治疗效果低下的主要因素是癌症的异质性。肿瘤发生的分子机制不明确,加上人体生理的复杂性,以及动物模型无法准确复制人体肿瘤微环境,这些都给新型癌症疗法的开发带来了巨大障碍。芯片上的肿瘤微环境(TMOC)是一个研究平台,它将三维细胞培养与微流控系统相结合,模拟了体内肿瘤微环境的基本成分和生理特征。它在芯片系统中提供了研究肿瘤进展的动态环境,可能预示着癌症研究的突破。在本综述中,我们将总结该平台目前的进展,包括各种类型的 TMOC 及其在不同类型癌症中的应用。从我们的角度来看,TMOC 平台需要加强与组织工程技术和微物理环境的整合,才能发展成为更完善的癌症研究临床前模型。
{"title":"Tumor-microenvironment-on-a-chip: the construction and application.","authors":"Hanzheng Xu, Jiangtao Wen, Jiahua Yang, Shufen Zhou, Yijie Li, Ke Xu, Wei Li, Sen Li","doi":"10.1186/s12964-024-01884-4","DOIUrl":"10.1186/s12964-024-01884-4","url":null,"abstract":"<p><p>Currently, despite the vast amounts of time and money invested in cancer treatment, cancer remains one of the primary threats to human life. The primary factor contributing to the low treatment efficacy is cancer heterogeneity. The unclear molecular mechanisms underlying tumorigenesis, coupled with the complexity of human physiology, and the inability of animal models to accurately replicate the human tumor microenvironment, pose significant hurdles in the development of novel cancer therapies. Tumor-microenvironment-on-chip (TMOC) represents a research platform that integrates three-dimensional cell culture with microfluidic systems, simulating the essential components and physiological traits of the in vivo tumor microenvironment. It offers a dynamic setting within the chip system to study tumor progression, potentially heralding a breakthrough in cancer research. In this review, we will summarize the current advancements in this platform, encompassing various types of TMOCs and their applications in different types of cancer. From our perspective, the TMOC platform necessitates enhanced integration with tissue engineering techniques and microphysiological environments before it can evolve into a more refined preclinical model for cancer research.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"515"},"PeriodicalIF":8.2,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515741/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142513371","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
Spatial-transcriptomic profiling: a new lens for understanding myelofibrosis pathophysiology. 空间转录组分析:了解骨髓纤维化病理生理学的新视角。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-21 DOI: 10.1186/s12964-024-01877-3
Edoardo Peroni, Elisabetta Calistri, Rosario Amato, Michele Gottardi, Antonio Rosato

Myelofibrosis (MF) is a complex myeloproliferative neoplasm characterized by abnormal hematopoietic stem cell proliferation and subsequent bone marrow (BM) fibrosis. First documented in the late 19th century, MF has since been extensively studied to unravel its pathophysiology, clinical phenotypes, and therapeutic interventions. MF can be classified into primary and secondary forms, both driven by mutations in genes such as JAK2, CALR, and MPL, which activate the JAK-STAT signaling pathway. These driver mutations are frequently accompanied by additional non-driver mutations in genes like TET2, SRSF2, and TP53, contributing to disease complexity. The BM microenvironment, consisting of stromal cells, extracellular matrix, and cytokines such as TGF-β and TNF-α, plays a critical role in fibrosis and aberrant hematopoiesis. Clinically, MF manifests with symptoms ranging from anemia, splenomegaly, and fatigue to severe complications such as leukemic transformation. Splenomegaly, caused by extramedullary hematopoiesis, leads to abdominal discomfort and early satiety. Current therapeutic strategies include JAK inhibitors like Ruxolitinib, which target the JAK-STAT pathway, alongside supportive treatments such as blood transfusions, erythropoiesis-stimulating agents and developing combinatorial approaches. Allogeneic hematopoietic stem cell transplantation remains the only curative option, though it is limited to younger, high-risk patients. Recently approved JAK inhibitors, including Fedratinib, Pacritinib, and Momelotinib, have expanded the therapeutic landscape. Spatially Resolved Transcriptomics (SRT) has revolutionized the study of gene expression within the spatial context of tissues, providing unprecedented insights into cellular heterogeneity, spatial gene regulation, and microenvironmental interactions, including stromal-hematopoietic dynamics. SRT enables high-resolution mapping of gene expression in the BM and spleen, revealing molecular signatures, spatial heterogeneity, and pathological niches that drive disease progression. These technologies elucidate the role of the spleen in MF, highlighting its transformation into a site of abnormal hematopoietic activity, fibrotic changes, and immune cell infiltration, functioning as a "tumor surrogate." By profiling diverse cell populations and molecular alterations within the BM and spleen, SRT facilitates a deeper understanding of MF pathophysiology, helping identify novel therapeutic targets and biomarkers. Ultimately, integrating spatial transcriptomics into MF research promises to enhance diagnostic precision and therapeutic innovation, addressing the multifaceted challenges of this disease.

骨髓纤维化(MF)是一种复杂的骨髓增生性肿瘤,其特点是造血干细胞异常增殖和随后的骨髓(BM)纤维化。骨髓纤维化最早见于 19 世纪末,此后人们对其进行了广泛研究,以揭示其病理生理学、临床表型和治疗干预措施。骨髓纤维化可分为原发性和继发性两种,均由激活 JAK-STAT 信号通路的 JAK2、CALR 和 MPL 等基因突变驱动。这些驱动基因突变常常伴随着 TET2、SRSF2 和 TP53 等基因的非驱动基因突变,从而导致疾病的复杂性。由基质细胞、细胞外基质和细胞因子(如 TGF-β 和 TNF-α)组成的 BM 微环境在纤维化和异常造血中起着至关重要的作用。在临床上,骨髓增生异常综合征的症状从贫血、脾肿大、乏力到白血病转化等严重并发症不等。髓外造血引起的脾肿大会导致腹部不适和早饱。目前的治疗策略包括以 JAK-STAT 通路为靶点的 JAK 抑制剂(如 Ruxolitinib),以及输血、促红细胞生成剂和开发组合方法等支持性治疗。异基因造血干细胞移植仍然是唯一的治疗选择,但仅限于年轻的高危患者。最近批准的 JAK 抑制剂,包括 Fedratinib、Pacritinib 和 Momelotinib,扩大了治疗范围。空间分辨转录组学(SRT)彻底改变了组织空间背景下的基因表达研究,为细胞异质性、空间基因调控和微环境相互作用(包括基质-造血动态)提供了前所未有的见解。SRT 能够高分辨率地绘制 BM 和脾脏中的基因表达图谱,揭示分子特征、空间异质性和驱动疾病进展的病理龛位。这些技术阐明了脾脏在骨髓纤维化中的作用,强调了脾脏转变为造血活动异常、纤维化变化和免疫细胞浸润的部位,起到了 "肿瘤替代物 "的作用。通过剖析骨髓和脾脏内不同的细胞群和分子变化,SRT 有助于加深对骨髓纤维化病理生理学的理解,帮助确定新的治疗靶点和生物标志物。最终,将空间转录组学融入骨髓纤维瘤研究有望提高诊断的精确性和治疗的创新性,从而应对该疾病的多方面挑战。
{"title":"Spatial-transcriptomic profiling: a new lens for understanding myelofibrosis pathophysiology.","authors":"Edoardo Peroni, Elisabetta Calistri, Rosario Amato, Michele Gottardi, Antonio Rosato","doi":"10.1186/s12964-024-01877-3","DOIUrl":"10.1186/s12964-024-01877-3","url":null,"abstract":"<p><p>Myelofibrosis (MF) is a complex myeloproliferative neoplasm characterized by abnormal hematopoietic stem cell proliferation and subsequent bone marrow (BM) fibrosis. First documented in the late 19th century, MF has since been extensively studied to unravel its pathophysiology, clinical phenotypes, and therapeutic interventions. MF can be classified into primary and secondary forms, both driven by mutations in genes such as JAK2, CALR, and MPL, which activate the JAK-STAT signaling pathway. These driver mutations are frequently accompanied by additional non-driver mutations in genes like TET2, SRSF2, and TP53, contributing to disease complexity. The BM microenvironment, consisting of stromal cells, extracellular matrix, and cytokines such as TGF-β and TNF-α, plays a critical role in fibrosis and aberrant hematopoiesis. Clinically, MF manifests with symptoms ranging from anemia, splenomegaly, and fatigue to severe complications such as leukemic transformation. Splenomegaly, caused by extramedullary hematopoiesis, leads to abdominal discomfort and early satiety. Current therapeutic strategies include JAK inhibitors like Ruxolitinib, which target the JAK-STAT pathway, alongside supportive treatments such as blood transfusions, erythropoiesis-stimulating agents and developing combinatorial approaches. Allogeneic hematopoietic stem cell transplantation remains the only curative option, though it is limited to younger, high-risk patients. Recently approved JAK inhibitors, including Fedratinib, Pacritinib, and Momelotinib, have expanded the therapeutic landscape. Spatially Resolved Transcriptomics (SRT) has revolutionized the study of gene expression within the spatial context of tissues, providing unprecedented insights into cellular heterogeneity, spatial gene regulation, and microenvironmental interactions, including stromal-hematopoietic dynamics. SRT enables high-resolution mapping of gene expression in the BM and spleen, revealing molecular signatures, spatial heterogeneity, and pathological niches that drive disease progression. These technologies elucidate the role of the spleen in MF, highlighting its transformation into a site of abnormal hematopoietic activity, fibrotic changes, and immune cell infiltration, functioning as a \"tumor surrogate.\" By profiling diverse cell populations and molecular alterations within the BM and spleen, SRT facilitates a deeper understanding of MF pathophysiology, helping identify novel therapeutic targets and biomarkers. Ultimately, integrating spatial transcriptomics into MF research promises to enhance diagnostic precision and therapeutic innovation, addressing the multifaceted challenges of this disease.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"510"},"PeriodicalIF":8.2,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492555/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481425","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
Gasdermin D promotes development of intestinal tumors through regulating IL-1β release and gut microbiota composition. Gasdermin D通过调节IL-1β的释放和肠道微生物群的组成促进肠道肿瘤的发展。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-21 DOI: 10.1186/s12964-024-01890-6
Hanchao Gao, Weilong Li, Shi Xu, Zigan Xu, Wenjun Hu, Litao Pan, Kewang Luo, Ting Xie, Yeye Yu, Huimin Sun, Liwen Huang, Peishan Chen, Jinmei Wu, Dexing Yang, Lian Li, Shaodong Luan, Mengtao Cao, Pengfei Chen

The interplay between gut microbiota and host is crucial for maintaining host health. When this balance is broken, various diseases can arise, including colorectal cancer (CRC). However, the mechanism by which gut microbiota and host interactions mediate CRC development remains unclear. Here, we found that Gasdermin D (GSDMD), an inflammasome effector responsible for forming membrane pores to mediate cell pyroptosis, was upregulated in both human and mouse intestinal tumor samples. GSDMD deficiency significantly suppressed intestinal tumor development in Apcmin/+ mice, a spontaneous CRC mouse model. Apcmin/+Gsdmd-/- mice exhibited reduced IL-1β release in the intestine, and the administration of recombinant mouse IL-1β partially restored intestinal tumor development in Apcmin/+Gsdmd-/- mice. Moreover, 16s rRNA sequencing showed a substantial increase in Lactobacillus abundance in the feces of Apcmin/+Gsdmd-/- mice compared to Apcmin/+ mice. Concurrently, Kynurenine (Kyn), a metabolite derived from host tryptophan (Trp) metabolism, was significantly decreased in the feces of Apcmin/+Gsdmd-/- mice, as shown by metabolite analysis. Additionally, Kyn levels were inversely correlated with Lactobacillus abundance. Furthermore, the administration of exogenous Kyn also promoted intestinal tumor development in Apcmin/+Gsdmd-/- mice. Thus, GSDMD promotes spontaneous CRC development through increasing IL-1β release and Kyn production. Our data suggest an association between GSDMD, gut microbiota, the host Trp/Kyn pathway, and CRC development.

肠道微生物群与宿主之间的相互作用对维持宿主健康至关重要。一旦这种平衡被打破,就会引发各种疾病,包括结直肠癌(CRC)。然而,肠道微生物群与宿主相互作用介导 CRC 发展的机制仍不清楚。在这里,我们发现人和小鼠肠道肿瘤样本中的Gasdermin D(GSDMD)上调,Gasdermin D是一种炎症小体效应物,负责形成膜孔以介导细胞热解。缺乏 GSDMD 能明显抑制 Apcmin/+ 小鼠(一种自发的 CRC 小鼠模型)肠道肿瘤的发展。Apcmin/+Gsdmd-/-小鼠肠道中的IL-1β释放减少,服用重组小鼠IL-1β可部分恢复Apcmin/+Gsdmd-/-小鼠的肠道肿瘤发生。此外,16s rRNA测序显示,与Apcmin/+小鼠相比,Apcmin/+Gsdmd-/-小鼠粪便中乳酸杆菌的数量大幅增加。同时,代谢物分析表明,Apcmin/+Gsdmd-/-小鼠粪便中的犬尿氨酸(Kyn)显著减少,犬尿氨酸是宿主色氨酸(Trp)代谢产生的一种代谢物。此外,Kyn 的水平与乳酸杆菌的丰度成反比。此外,给予外源性 Kyn 还会促进 Apcmin/+Gsdmd-/- 小鼠肠道肿瘤的发展。因此,GSDMD 通过增加 IL-1β 的释放和 Kyn 的产生来促进自发性 CRC 的发展。我们的数据表明,GSDMD、肠道微生物群、宿主 Trp/Kyn 通路和 CRC 发育之间存在关联。
{"title":"Gasdermin D promotes development of intestinal tumors through regulating IL-1β release and gut microbiota composition.","authors":"Hanchao Gao, Weilong Li, Shi Xu, Zigan Xu, Wenjun Hu, Litao Pan, Kewang Luo, Ting Xie, Yeye Yu, Huimin Sun, Liwen Huang, Peishan Chen, Jinmei Wu, Dexing Yang, Lian Li, Shaodong Luan, Mengtao Cao, Pengfei Chen","doi":"10.1186/s12964-024-01890-6","DOIUrl":"10.1186/s12964-024-01890-6","url":null,"abstract":"<p><p>The interplay between gut microbiota and host is crucial for maintaining host health. When this balance is broken, various diseases can arise, including colorectal cancer (CRC). However, the mechanism by which gut microbiota and host interactions mediate CRC development remains unclear. Here, we found that Gasdermin D (GSDMD), an inflammasome effector responsible for forming membrane pores to mediate cell pyroptosis, was upregulated in both human and mouse intestinal tumor samples. GSDMD deficiency significantly suppressed intestinal tumor development in Apc<sup>min/+</sup> mice, a spontaneous CRC mouse model. Apc<sup>min/+</sup>Gsdmd<sup>-/-</sup> mice exhibited reduced IL-1β release in the intestine, and the administration of recombinant mouse IL-1β partially restored intestinal tumor development in Apc<sup>min/+</sup>Gsdmd<sup>-/-</sup> mice. Moreover, 16s rRNA sequencing showed a substantial increase in Lactobacillus abundance in the feces of Apc<sup>min/+</sup>Gsdmd<sup>-/-</sup> mice compared to Apc<sup>min/+</sup> mice. Concurrently, Kynurenine (Kyn), a metabolite derived from host tryptophan (Trp) metabolism, was significantly decreased in the feces of Apc<sup>min/+</sup>Gsdmd<sup>-/-</sup> mice, as shown by metabolite analysis. Additionally, Kyn levels were inversely correlated with Lactobacillus abundance. Furthermore, the administration of exogenous Kyn also promoted intestinal tumor development in Apc<sup>min/+</sup>Gsdmd<sup>-/-</sup> mice. Thus, GSDMD promotes spontaneous CRC development through increasing IL-1β release and Kyn production. Our data suggest an association between GSDMD, gut microbiota, the host Trp/Kyn pathway, and CRC development.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"511"},"PeriodicalIF":8.2,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492562/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481415","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
Oncolytic avian reovirus-sensitized tumor infiltrating CD8+ T cells triggering immunogenic apoptosis in gastric cancer. 肿瘤溶解性禽再病毒致敏的肿瘤浸润性 CD8+ T 细胞引发胃癌免疫性凋亡。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-21 DOI: 10.1186/s12964-024-01888-0
Yi-Ying Wu, Feng-Hsu Wu, I-Chun Chen, Tsai-Ling Liao, Muhammad Munir, Hung-Jen Liu

Background: Gastric cancer (GC) is a leading malignant disease in numerous countries, including Taiwan with limited therapeutic options. Animal viruses including oncolytic avian reovirus (ARV) have the possibility to avoid pre-existing immunity in humans, while being safe and immunostimulatory. Here, we provide a novel insight into oncolytic ARV and UV-ARV-sensitized patient's peripheral blood mononuclear cells (P-PBMCs) and tumor infiltrating lymphocytes (TILs) killing primary GC (PGC) cells through the surface TLR3 and TRAIL/DR4/DR5 immunogenic apoptosis pathway.

Methods: We conducted a comprehensive study to reveal whether ARV- or UV-inactivated ARV (UV-ARV)-modulated P-PBMCs or TILs killing ARV- and UV-ARV-sensitized AGS cells and PGC cells derived from clinical patients and to investigate the regulation of surface TLR3 receptor and upstream signaling pathways. Apoptosis analysis by flow cytometry and Western blot, suppression of signal pathway by specific inhibitors, in situ proximity ligation assay (PLA), time-resolved flurometry and lactate dehydrogenase (LDH) cytotoxicity assays, and an in vitro co-culture model were established to study the interplay between ARV- and UV-ARV-sensitized P-PBMCs and TILs to kill PGC cells and their upstream pathways.

Results: Our results reveal that increased levels of DR4 and DR5 were observed in ARV and UV-ARV sensitized PGC cells through the TLR3/p38/p53 signaling pathway. Importantly, we found that the σC protein of ARV or UV-ARV interacted with surface TLR3 of CD8+ TILs, thereby triggering the TLR3/NF-κB/IFN-γ/TRAIL signaling pathway which induces immunogenic apoptosis of PGC cells. This study sheds further light on the molecular basis behind ARV oncolysis and facilitates the ARV or UV-ARV as a cancer therapeutic.

Conclusions: The study provides novel insights into ARV- or UV-ARV-sensitized P-PBMCs and CD8+ TILs to kill PGC cells through the immunogenic apoptosis pathway. We conclude that P-PBMCs can easily be obtained from GC patients and provide a rich source as TILs to kill PGC cells.

背景胃癌(GC)是包括台湾在内的许多国家的主要恶性疾病,但治疗方案有限。包括溶瘤禽再病毒(ARV)在内的动物病毒可以避免人类原有的免疫力,同时具有安全性和免疫刺激作用。在此,我们对溶瘤ARV和UV-ARV致敏的患者外周血单核细胞(P-PBMCs)和肿瘤浸润淋巴细胞(TILs)通过表面TLR3和TRAIL/DR4/DR5免疫原性凋亡途径杀死原发性GC(PGC)细胞进行了新的深入研究:我们进行了一项综合研究,以揭示ARV或UV灭活ARV(UV-ARV)调控的P-PBMCs或TILs是否杀伤ARV和UV-ARV致敏的AGS细胞和来自临床患者的PGC细胞,并研究表面TLR3受体和上游信号通路的调控。通过流式细胞术和Western印迹进行细胞凋亡分析、用特异性抑制剂抑制信号通路、原位近接试验(PLA)、时间分辨荧光测定法和乳酸脱氢酶(LDH)细胞毒性试验以及体外共培养模型,研究了ARV和UV-ARV致敏的P-PBMCs和TILs杀死PGC细胞的相互作用及其上游通路:结果:我们的研究结果表明,通过TLR3/p38/p53信号通路,在ARV和UV-ARV致敏的PGC细胞中观察到DR4和DR5水平升高。重要的是,我们发现 ARV 或 UV-ARV 的 σC 蛋白与 CD8+ TILs 表面的 TLR3 相互作用,从而触发 TLR3/NF-κB/IFN-γ/TRAIL 信号通路,诱导 PGC 细胞免疫性凋亡。这项研究进一步揭示了ARV溶瘤背后的分子基础,有助于将ARV或UV-ARV作为癌症疗法:本研究为ARV或UV-ARV致敏的P-PBMCs和CD8+ TILs通过免疫原性凋亡途径杀死PGC细胞提供了新的见解。我们得出的结论是,P-PBMCs 可以很容易地从 GC 患者身上获得,并为 TILs 提供了丰富的来源,从而杀死 PGC 细胞。
{"title":"Oncolytic avian reovirus-sensitized tumor infiltrating CD8<sup>+</sup> T cells triggering immunogenic apoptosis in gastric cancer.","authors":"Yi-Ying Wu, Feng-Hsu Wu, I-Chun Chen, Tsai-Ling Liao, Muhammad Munir, Hung-Jen Liu","doi":"10.1186/s12964-024-01888-0","DOIUrl":"10.1186/s12964-024-01888-0","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) is a leading malignant disease in numerous countries, including Taiwan with limited therapeutic options. Animal viruses including oncolytic avian reovirus (ARV) have the possibility to avoid pre-existing immunity in humans, while being safe and immunostimulatory. Here, we provide a novel insight into oncolytic ARV and UV-ARV-sensitized patient's peripheral blood mononuclear cells (P-PBMCs) and tumor infiltrating lymphocytes (TILs) killing primary GC (PGC) cells through the surface TLR3 and TRAIL/DR4/DR5 immunogenic apoptosis pathway.</p><p><strong>Methods: </strong>We conducted a comprehensive study to reveal whether ARV- or UV-inactivated ARV (UV-ARV)-modulated P-PBMCs or TILs killing ARV- and UV-ARV-sensitized AGS cells and PGC cells derived from clinical patients and to investigate the regulation of surface TLR3 receptor and upstream signaling pathways. Apoptosis analysis by flow cytometry and Western blot, suppression of signal pathway by specific inhibitors, in situ proximity ligation assay (PLA), time-resolved flurometry and lactate dehydrogenase (LDH) cytotoxicity assays, and an in vitro co-culture model were established to study the interplay between ARV- and UV-ARV-sensitized P-PBMCs and TILs to kill PGC cells and their upstream pathways.</p><p><strong>Results: </strong>Our results reveal that increased levels of DR4 and DR5 were observed in ARV and UV-ARV sensitized PGC cells through the TLR3/p38/p53 signaling pathway. Importantly, we found that the σC protein of ARV or UV-ARV interacted with surface TLR3 of CD8<sup>+</sup> TILs, thereby triggering the TLR3/NF-κB/IFN-γ/TRAIL signaling pathway which induces immunogenic apoptosis of PGC cells. This study sheds further light on the molecular basis behind ARV oncolysis and facilitates the ARV or UV-ARV as a cancer therapeutic.</p><p><strong>Conclusions: </strong>The study provides novel insights into ARV- or UV-ARV-sensitized P-PBMCs and CD8<sup>+</sup> TILs to kill PGC cells through the immunogenic apoptosis pathway. We conclude that P-PBMCs can easily be obtained from GC patients and provide a rich source as TILs to kill PGC cells.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"514"},"PeriodicalIF":8.2,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11494775/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481423","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
The signature of extracellular vesicles in hypoxic breast cancer and their therapeutic engineering. 缺氧性乳腺癌细胞外囊泡的特征及其治疗工程。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-21 DOI: 10.1186/s12964-024-01870-w
Baiheng Zhu, Kehao Xiang, Tanghua Li, Xin Li, Fujun Shi

Breast cancer (BC) currently ranks second in the global cancer incidence rate. Hypoxia is a common phenomenon in BC. Under hypoxic conditions, cells in the tumor microenvironment (TME) secrete numerous extracellular vesicles (EVs) to achieve intercellular communication and alter the metabolism of primary and metastatic tumors that shape the TME. In addition, emerging studies have indicated that hypoxia can promote resistance to tumor treatment. Engineered EVs are expected to become carriers for cancer treatment due to their high biocompatibility, low immunogenicity, high drug delivery efficiency, and ease of modification. In this review, we summarize the mechanisms of EVs in the primary TME and distant metastasis of BC under hypoxic conditions. Additionally, we highlight the potential applications of engineered EVs in mitigating the malignant phenotypes of BC cells under hypoxia.

乳腺癌(BC)目前在全球癌症发病率中排名第二。缺氧是乳腺癌的常见现象。在缺氧条件下,肿瘤微环境(TME)中的细胞会分泌大量细胞外囊泡(EVs),以实现细胞间的交流,并改变形成 TME 的原发性和转移性肿瘤的新陈代谢。此外,新的研究表明,缺氧会促进肿瘤治疗的抗药性。工程EV具有生物相容性高、免疫原性低、给药效率高、易于改造等特点,有望成为癌症治疗的载体。在这篇综述中,我们总结了在缺氧条件下,EVs 在原发性 TME 和 BC 远处转移中的作用机制。此外,我们还强调了工程EVs在缺氧条件下减轻BC细胞恶性表型的潜在应用。
{"title":"The signature of extracellular vesicles in hypoxic breast cancer and their therapeutic engineering.","authors":"Baiheng Zhu, Kehao Xiang, Tanghua Li, Xin Li, Fujun Shi","doi":"10.1186/s12964-024-01870-w","DOIUrl":"10.1186/s12964-024-01870-w","url":null,"abstract":"<p><p>Breast cancer (BC) currently ranks second in the global cancer incidence rate. Hypoxia is a common phenomenon in BC. Under hypoxic conditions, cells in the tumor microenvironment (TME) secrete numerous extracellular vesicles (EVs) to achieve intercellular communication and alter the metabolism of primary and metastatic tumors that shape the TME. In addition, emerging studies have indicated that hypoxia can promote resistance to tumor treatment. Engineered EVs are expected to become carriers for cancer treatment due to their high biocompatibility, low immunogenicity, high drug delivery efficiency, and ease of modification. In this review, we summarize the mechanisms of EVs in the primary TME and distant metastasis of BC under hypoxic conditions. Additionally, we highlight the potential applications of engineered EVs in mitigating the malignant phenotypes of BC cells under hypoxia.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"512"},"PeriodicalIF":8.2,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492701/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481428","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
Decoding the epitranscriptome: a new frontier for cancer therapy and drug resistance. 解码表观转录组:癌症治疗和抗药性的新领域。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-21 DOI: 10.1186/s12964-024-01854-w
Lu Tang, Hua Tian, Qi Min, Huili You, Mengshuang Yin, Liqiong Yang, Yueshui Zhao, Xu Wu, Mingxing Li, Fukuan Du, Yu Chen, Shuai Deng, Xiaobing Li, Meijuan Chen, Li Gu, Yuhong Sun, Zhangang Xiao, Wanping Li, Jing Shen

As the role of RNA modification in gene expression regulation and human diseases, the "epitranscriptome" has been shown to be an important player in regulating many physiological and pathological processes. Meanwhile, the phenomenon of cancer drug resistance is becoming more and more frequent, especially in the case of cancer chemotherapy resistance. In recent years, research on relationship between post-transcriptional modification and cancer including drug resistance has become a hot topic, especially the methylation of the sixth nitrogen site of RNA adenosine-m6A (N6-methyladenosine). m6A modification is the most common post-transcriptional modification of eukaryotic mRNA, accounting for 80% of RNA methylation modifications. At the same time, several other modifications of RNA, such as N1-methyladenosine (m1A), 5-methylcytosine (m5C), 3-methylcytosine (m3C), pseudouridine (Ψ) and N7-methylguanosine (m7G) have also been demonstrated to be involved in cancer and drug resistance. This review mainly discusses the research progress of RNA modifications in the field of cancer and drug resistance and targeting of m6A regulators by small molecule modulators, providing reference for future study and development of combination therapy to reverse cancer drug resistance.

由于 RNA 修饰在基因表达调控和人类疾病中的作用,"表观转录组 "已被证明是调控许多生理和病理过程的重要角色。与此同时,癌症耐药现象日益频繁,尤其是癌症化疗耐药。近年来,转录后修饰与癌症(包括耐药性)之间关系的研究成为热点,尤其是 RNA 第六个氮位点腺苷-m6A(N6-甲基腺苷)的甲基化。与此同时,其他一些 RNA 甲基化修饰,如 N1-甲基腺苷(m1A)、5-甲基胞嘧啶(m5C)、3-甲基胞嘧啶(m3C)、假尿苷(Ψ)和 N7-甲基鸟苷(m7G)也被证实与癌症和耐药性有关。这篇综述主要讨论了 RNA 修饰在癌症和耐药性领域的研究进展,以及小分子调节剂靶向 m6A 调节剂的情况,为今后研究和开发逆转癌症耐药性的联合疗法提供参考。
{"title":"Decoding the epitranscriptome: a new frontier for cancer therapy and drug resistance.","authors":"Lu Tang, Hua Tian, Qi Min, Huili You, Mengshuang Yin, Liqiong Yang, Yueshui Zhao, Xu Wu, Mingxing Li, Fukuan Du, Yu Chen, Shuai Deng, Xiaobing Li, Meijuan Chen, Li Gu, Yuhong Sun, Zhangang Xiao, Wanping Li, Jing Shen","doi":"10.1186/s12964-024-01854-w","DOIUrl":"10.1186/s12964-024-01854-w","url":null,"abstract":"<p><p>As the role of RNA modification in gene expression regulation and human diseases, the \"epitranscriptome\" has been shown to be an important player in regulating many physiological and pathological processes. Meanwhile, the phenomenon of cancer drug resistance is becoming more and more frequent, especially in the case of cancer chemotherapy resistance. In recent years, research on relationship between post-transcriptional modification and cancer including drug resistance has become a hot topic, especially the methylation of the sixth nitrogen site of RNA adenosine-m<sup>6</sup>A (N6-methyladenosine). m<sup>6</sup>A modification is the most common post-transcriptional modification of eukaryotic mRNA, accounting for 80% of RNA methylation modifications. At the same time, several other modifications of RNA, such as N1-methyladenosine (m<sup>1</sup>A), 5-methylcytosine (m<sup>5</sup>C), 3-methylcytosine (m<sup>3</sup>C), pseudouridine (Ψ) and N7-methylguanosine (m<sup>7</sup>G) have also been demonstrated to be involved in cancer and drug resistance. This review mainly discusses the research progress of RNA modifications in the field of cancer and drug resistance and targeting of m<sup>6</sup>A regulators by small molecule modulators, providing reference for future study and development of combination therapy to reverse cancer drug resistance.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"513"},"PeriodicalIF":8.2,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492518/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481413","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
Lanthanide conjugate Pr-MPO elicits anti-cancer activity by targeting lysosomal machinery and inducing zinc-dependent cataplerosis. 镧系元素共轭物Pr-MPO通过靶向溶酶体机制和诱导锌依赖性催化反应而激发抗癌活性。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-19 DOI: 10.1186/s12964-024-01883-5
Gregory Lucien Bellot, Dan Liu, Marc Fivaz, Sanjiv K Yadav, Charanjit Kaur, Shazib Pervaiz

Acquired drug resistance is a major challenge in the management of cancer, which underscores the need for discovery and development of novel therapeutic strategies. We report here the mechanism of the anti-cancer activity of a small coordinate complex composed of the rare earth metal praseodymium (Pr) and mercaptopyridine oxide (MPO; pyrithione). Exposure of cancer cells to relatively low concentrations of the conjugate Pr-MPO (5 µM) significantly impairs cell survival in a p53-independent manner and irrespective of the drug resistant phenotype. Mechanistically, Pr-MPO-induced cell death is caspase-independent, not inhibitable by necrostatin, but associated with the appearance of autophagy markers. However, further analysis revealed incomplete autophagic flux, thus suggesting altered integrity of lysosomal machinery. Supporting the lysosomal targeting activity are data demonstrating increased lysosomal Ca2+ accumulation and alkalinization, which coincides with cytosolic acidification (drop in pHc from 7.75 to 7.00). In parallel, an increase in lysosomal activity of glycosidase alpha acid (GAA), involved in passive glycogen breakdown, correlates with rapid depletion of glucose stores upon Pr-MPO treatment. This is associated with swift cataplerosis of TCA cycle intermediates, loss of NAD+/NADH and increase in pyruvate dehydrogenase (PDH) activity to compensate for pyruvate loss. Addition of exogenous pyruvate rescued cell survival. Notably, lysosomal impairment and metabolic catastrophe triggered by Pr-MPO are suggestive of Zn2+-mediated cytotoxicity, which is confirmed by the ability of Zn2+ chelator TPEN to block Pr-MPO-mediated anti-tumor activity. Together, these results highlight the ability of the small molecule lanthanide conjugate to target the cells' waste clearing machinery as well as mitochondrial metabolism for Zn2+-mediated execution of cancer cells, which could have therapeutic potential against cancers with high metabolic activity.

获得性耐药性是治疗癌症的一大挑战,这凸显了发现和开发新型治疗策略的必要性。我们在此报告由稀土金属镨(Pr)和巯基吡啶氧化物(MPO;吡硫鎓)组成的小配位复合物的抗癌活性机制。将癌细胞暴露于相对低浓度的轭合物 Pr-MPO(5 µM)中,会以不依赖 p53 的方式显著降低细胞的存活率,且与抗药性表型无关。从机理上讲,Pr-MPO 诱导的细胞死亡与 Caspase 无关,不受坏死素的抑制,但与自噬标记物的出现有关。然而,进一步分析发现自噬通量不完全,这表明溶酶体机制的完整性发生了改变。支持溶酶体靶向活动的数据显示,溶酶体 Ca2+ 积累和碱化增加,与细胞酸化(pHc 从 7.75 降至 7.00)同时发生。与此同时,溶酶体中参与被动糖原分解的α-酸糖苷酶(GAA)活性的增加与 Pr-MPO 处理后葡萄糖储存的快速耗竭有关。这与 TCA 循环中间产物的快速催化、NAD+/NADH 的损失和丙酮酸脱氢酶(PDH)活性的增加有关,以补偿丙酮酸的损失。加入外源丙酮酸可挽救细胞的存活。值得注意的是,Pr-MPO 引发的溶酶体损伤和代谢灾难表明了 Zn2+ 介导的细胞毒性,而 Zn2+ 螯合剂 TPEN 阻断 Pr-MPO 介导的抗肿瘤活性的能力也证实了这一点。这些结果突出表明,小分子镧系元素轭合物能够靶向细胞的废物清除机制和线粒体代谢,从而通过 Zn2+ 介导的作用杀死癌细胞,这对具有高代谢活性的癌症具有治疗潜力。
{"title":"Lanthanide conjugate Pr-MPO elicits anti-cancer activity by targeting lysosomal machinery and inducing zinc-dependent cataplerosis.","authors":"Gregory Lucien Bellot, Dan Liu, Marc Fivaz, Sanjiv K Yadav, Charanjit Kaur, Shazib Pervaiz","doi":"10.1186/s12964-024-01883-5","DOIUrl":"10.1186/s12964-024-01883-5","url":null,"abstract":"<p><p>Acquired drug resistance is a major challenge in the management of cancer, which underscores the need for discovery and development of novel therapeutic strategies. We report here the mechanism of the anti-cancer activity of a small coordinate complex composed of the rare earth metal praseodymium (Pr) and mercaptopyridine oxide (MPO; pyrithione). Exposure of cancer cells to relatively low concentrations of the conjugate Pr-MPO (5 µM) significantly impairs cell survival in a p53-independent manner and irrespective of the drug resistant phenotype. Mechanistically, Pr-MPO-induced cell death is caspase-independent, not inhibitable by necrostatin, but associated with the appearance of autophagy markers. However, further analysis revealed incomplete autophagic flux, thus suggesting altered integrity of lysosomal machinery. Supporting the lysosomal targeting activity are data demonstrating increased lysosomal Ca<sup>2+</sup> accumulation and alkalinization, which coincides with cytosolic acidification (drop in pH<sub>c</sub> from 7.75 to 7.00). In parallel, an increase in lysosomal activity of glycosidase alpha acid (GAA), involved in passive glycogen breakdown, correlates with rapid depletion of glucose stores upon Pr-MPO treatment. This is associated with swift cataplerosis of TCA cycle intermediates, loss of NAD<sup>+</sup>/NADH and increase in pyruvate dehydrogenase (PDH) activity to compensate for pyruvate loss. Addition of exogenous pyruvate rescued cell survival. Notably, lysosomal impairment and metabolic catastrophe triggered by Pr-MPO are suggestive of Zn<sup>2+</sup>-mediated cytotoxicity, which is confirmed by the ability of Zn<sup>2+</sup> chelator TPEN to block Pr-MPO-mediated anti-tumor activity. Together, these results highlight the ability of the small molecule lanthanide conjugate to target the cells' waste clearing machinery as well as mitochondrial metabolism for Zn<sup>2+</sup>-mediated execution of cancer cells, which could have therapeutic potential against cancers with high metabolic activity.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"509"},"PeriodicalIF":8.2,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11490180/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481420","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
Alternative splicing in ovarian cancer. 卵巢癌中的替代剪接。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-18 DOI: 10.1186/s12964-024-01880-8
Liwei Wei, Yisheng Li, Jiawang Chen, Yuanmei Wang, Jianmin Wu, Huanming Yang, Yi Zhang

Ovarian cancer is the second leading cause of gynecologic cancer death worldwide, with only 20% of cases detected early due to its elusive nature, limiting successful treatment. Most deaths occur from the disease progressing to advanced stages. Despite advances in chemo- and immunotherapy, the 5-year survival remains below 50% due to high recurrence and chemoresistance. Therefore, leveraging new research perspectives to understand molecular signatures and identify novel therapeutic targets is crucial for improving the clinical outcomes of ovarian cancer. Alternative splicing, a fundamental mechanism of post-transcriptional gene regulation, significantly contributes to heightened genomic complexity and protein diversity. Increased awareness has emerged about the multifaceted roles of alternative splicing in ovarian cancer, including cell proliferation, metastasis, apoptosis, immune evasion, and chemoresistance. We begin with an overview of altered splicing machinery, highlighting increased expression of spliceosome components and associated splicing factors like BUD31, SF3B4, and CTNNBL1, and their relationships to ovarian cancer. Next, we summarize the impact of specific variants of CD44, ECM1, and KAI1 on tumorigenesis and drug resistance through diverse mechanisms. Recent genomic and bioinformatics advances have enhanced our understanding. By incorporating data from The Cancer Genome Atlas RNA-seq, along with clinical information, a series of prognostic models have been developed, which provided deeper insights into how the splicing influences prognosis, overall survival, the immune microenvironment, and drug sensitivity and resistance in ovarian cancer patients. Notably, novel splicing events, such as PIGV|1299|AP and FLT3LG|50,941|AP, have been identified in multiple prognostic models and are associated with poorer and improved prognosis, respectively. These novel splicing variants warrant further functional characterization to unlock the underlying molecular mechanisms. Additionally, experimental evidence has underscored the potential therapeutic utility of targeting alternative splicing events, exemplified by the observation that knockdown of splicing factor BUD31 or antisense oligonucleotide-induced BCL2L12 exon skipping promotes apoptosis of ovarian cancer cells. In clinical settings, bevacizumab, a humanized monoclonal antibody that specifically targets the VEGF-A isoform, has demonstrated beneficial effects in the treatment of patients with advanced epithelial ovarian cancer. In conclusion, this review constitutes the first comprehensive and detailed exposition of the intricate interplay between alternative splicing and ovarian cancer, underscoring the significance of alternative splicing events as pivotal determinants in cancer biology and as promising avenues for future diagnostic and therapeutic intervention.

卵巢癌是全球第二大妇科癌症死因,由于其难以捉摸的特性,只有 20% 的病例能被早期发现,从而限制了治疗的成功率。大多数患者死于疾病进展到晚期。尽管化疗和免疫疗法取得了进展,但由于高复发率和化疗耐药性,5 年生存率仍低于 50%。因此,利用新的研究视角来了解分子特征并确定新的治疗靶点,对于改善卵巢癌的临床治疗效果至关重要。替代剪接是转录后基因调控的基本机制,它极大地增加了基因组的复杂性和蛋白质的多样性。人们越来越意识到替代剪接在卵巢癌中的多方面作用,包括细胞增殖、转移、凋亡、免疫逃避和化疗抗性。我们首先概述了剪接机制的改变,强调了剪接体成分和相关剪接因子(如 BUD31、SF3B4 和 CTNNBL1)表达的增加及其与卵巢癌的关系。接下来,我们总结了 CD44、ECM1 和 KAI1 的特定变体通过不同机制对肿瘤发生和耐药性的影响。基因组学和生物信息学的最新进展加深了我们的理解。通过结合癌症基因组图谱 RNA-seq 数据和临床信息,我们建立了一系列预后模型,从而更深入地了解了剪接如何影响卵巢癌患者的预后、总体生存、免疫微环境以及药物敏感性和耐药性。值得注意的是,PIGV|1299|AP和FLT3LG|50,941|AP等新型剪接事件已在多个预后模型中被发现,并分别与预后较差和预后改善相关。这些新型剪接变异需要进一步的功能表征,以揭示其潜在的分子机制。此外,实验证据强调了靶向替代剪接事件的潜在治疗作用,例如,观察到剪接因子 BUD31 的敲除或反义寡核苷酸诱导的 BCL2L12 外显子跳越可促进卵巢癌细胞的凋亡。在临床上,贝伐珠单抗是一种特异性靶向血管内皮生长因子-A 同工酶的人源化单克隆抗体,在治疗晚期上皮性卵巢癌患者方面显示出有益的效果。总之,这篇综述首次全面而详细地阐述了替代剪接与卵巢癌之间错综复杂的相互作用,强调了替代剪接事件作为癌症生物学关键决定因素的重要意义,以及作为未来诊断和治疗干预手段的前景。
{"title":"Alternative splicing in ovarian cancer.","authors":"Liwei Wei, Yisheng Li, Jiawang Chen, Yuanmei Wang, Jianmin Wu, Huanming Yang, Yi Zhang","doi":"10.1186/s12964-024-01880-8","DOIUrl":"https://doi.org/10.1186/s12964-024-01880-8","url":null,"abstract":"<p><p>Ovarian cancer is the second leading cause of gynecologic cancer death worldwide, with only 20% of cases detected early due to its elusive nature, limiting successful treatment. Most deaths occur from the disease progressing to advanced stages. Despite advances in chemo- and immunotherapy, the 5-year survival remains below 50% due to high recurrence and chemoresistance. Therefore, leveraging new research perspectives to understand molecular signatures and identify novel therapeutic targets is crucial for improving the clinical outcomes of ovarian cancer. Alternative splicing, a fundamental mechanism of post-transcriptional gene regulation, significantly contributes to heightened genomic complexity and protein diversity. Increased awareness has emerged about the multifaceted roles of alternative splicing in ovarian cancer, including cell proliferation, metastasis, apoptosis, immune evasion, and chemoresistance. We begin with an overview of altered splicing machinery, highlighting increased expression of spliceosome components and associated splicing factors like BUD31, SF3B4, and CTNNBL1, and their relationships to ovarian cancer. Next, we summarize the impact of specific variants of CD44, ECM1, and KAI1 on tumorigenesis and drug resistance through diverse mechanisms. Recent genomic and bioinformatics advances have enhanced our understanding. By incorporating data from The Cancer Genome Atlas RNA-seq, along with clinical information, a series of prognostic models have been developed, which provided deeper insights into how the splicing influences prognosis, overall survival, the immune microenvironment, and drug sensitivity and resistance in ovarian cancer patients. Notably, novel splicing events, such as PIGV|1299|AP and FLT3LG|50,941|AP, have been identified in multiple prognostic models and are associated with poorer and improved prognosis, respectively. These novel splicing variants warrant further functional characterization to unlock the underlying molecular mechanisms. Additionally, experimental evidence has underscored the potential therapeutic utility of targeting alternative splicing events, exemplified by the observation that knockdown of splicing factor BUD31 or antisense oligonucleotide-induced BCL2L12 exon skipping promotes apoptosis of ovarian cancer cells. In clinical settings, bevacizumab, a humanized monoclonal antibody that specifically targets the VEGF-A isoform, has demonstrated beneficial effects in the treatment of patients with advanced epithelial ovarian cancer. In conclusion, this review constitutes the first comprehensive and detailed exposition of the intricate interplay between alternative splicing and ovarian cancer, underscoring the significance of alternative splicing events as pivotal determinants in cancer biology and as promising avenues for future diagnostic and therapeutic intervention.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"507"},"PeriodicalIF":8.2,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11488268/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481408","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
Insights into ubiquitinome dynamics in the host‒pathogen interplay during Francisella novicida infection. 洞察新弗朗西斯菌感染过程中宿主-病原体相互作用的泛素组动态。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-18 DOI: 10.1186/s12964-024-01887-1
Luyu Yang, Yanfeng Li, Qingqing Xie, Tao Xu, Xiaopeng Qi

Ubiquitination functions as an important posttranslational modification for orchestrating inflammatory immune responses and cell death during pathogenic infection. The ubiquitination machinery is a major target hijacked by pathogenic bacteria to promote their survival and proliferation. Type I interferon (IFN-I) plays detrimental roles in host defense against Francisella novicida (F. novicida) infection. The effects of IFN-I on the ubiquitination of host proteins during F. novicida infection remain unclear. Herein, we delineate the dynamic ubiquitinome alterations in both wild-type (WT) and interferon-alpha receptor-deficient (Ifnar-/-) primary bone marrow-derived macrophages (BMDMs) during F. novicida infection. Using diGly proteomics and stable isotope labeling (SILAC), we quantified ubiquitination sites in proteins from primary WT and Ifnar-/- BMDMs with and without F. novicida infection. Our mass spectrometry analysis identified 2,491 ubiquitination sites in 1,077 endogenous proteins. Our study revealed that F. novicida infection induces dynamic changes in the ubiquitination of proteins involved in the cell death, phagocytosis, and inflammatory response pathways. IFN-I signaling is essential for both the increase and reduction in ubiquitination in response to F. novicida infection. We identified IFN-I-dependent ubiquitination in proteins involved in glycolysis and vesicle transport processes and highlighted key hub proteins modified by ubiquitination within cell death pathways. These findings underscore the significant influence of IFN-I signaling on modulating ubiquitination during F. novicida infection and provide valuable insights into the complex interplay between the host and F. novicida.

泛素化是病原体感染期间协调炎症免疫反应和细胞死亡的一种重要的翻译后修饰。泛素化机制是病原细菌为促进其生存和增殖而劫持的一个主要目标。I 型干扰素(IFN-I)在宿主防御诺维茨基弗朗西斯菌(F. novicida)感染的过程中发挥着有害作用。IFN-I 对 F. novicida 感染期间宿主蛋白质泛素化的影响仍不清楚。在此,我们描述了野生型(WT)和干扰素-α受体缺陷型(Ifnar-/-)原发性骨髓源性巨噬细胞(BMDMs)在F. novicida感染期间泛素组的动态变化。利用 diGly 蛋白组学和稳定同位素标记(SILAC)技术,我们量化了 WT 和 Ifnar-/- BMDMs 在感染 F. novicida 和未感染 F. novicida 时的蛋白质泛素化位点。我们的质谱分析在 1,077 个内源蛋白质中发现了 2,491 个泛素化位点。我们的研究发现,F. novicida 感染会诱导参与细胞死亡、吞噬和炎症反应途径的蛋白质泛素化发生动态变化。IFN-I信号对于泛素化的增加和减少都是必不可少的。我们在参与糖酵解和囊泡运输过程的蛋白质中发现了 IFN-I 依赖性泛素化,并强调了细胞死亡途径中泛素化修饰的关键枢纽蛋白。这些发现强调了 IFN-I 信号在诺维卡氏酵母菌感染过程中对泛素化调节的重要影响,并为了解宿主与诺维卡氏酵母菌之间复杂的相互作用提供了宝贵的见解。
{"title":"Insights into ubiquitinome dynamics in the host‒pathogen interplay during Francisella novicida infection.","authors":"Luyu Yang, Yanfeng Li, Qingqing Xie, Tao Xu, Xiaopeng Qi","doi":"10.1186/s12964-024-01887-1","DOIUrl":"https://doi.org/10.1186/s12964-024-01887-1","url":null,"abstract":"<p><p>Ubiquitination functions as an important posttranslational modification for orchestrating inflammatory immune responses and cell death during pathogenic infection. The ubiquitination machinery is a major target hijacked by pathogenic bacteria to promote their survival and proliferation. Type I interferon (IFN-I) plays detrimental roles in host defense against Francisella novicida (F. novicida) infection. The effects of IFN-I on the ubiquitination of host proteins during F. novicida infection remain unclear. Herein, we delineate the dynamic ubiquitinome alterations in both wild-type (WT) and interferon-alpha receptor-deficient (Ifnar<sup>-/-</sup>) primary bone marrow-derived macrophages (BMDMs) during F. novicida infection. Using diGly proteomics and stable isotope labeling (SILAC), we quantified ubiquitination sites in proteins from primary WT and Ifnar<sup>-/-</sup> BMDMs with and without F. novicida infection. Our mass spectrometry analysis identified 2,491 ubiquitination sites in 1,077 endogenous proteins. Our study revealed that F. novicida infection induces dynamic changes in the ubiquitination of proteins involved in the cell death, phagocytosis, and inflammatory response pathways. IFN-I signaling is essential for both the increase and reduction in ubiquitination in response to F. novicida infection. We identified IFN-I-dependent ubiquitination in proteins involved in glycolysis and vesicle transport processes and highlighted key hub proteins modified by ubiquitination within cell death pathways. These findings underscore the significant influence of IFN-I signaling on modulating ubiquitination during F. novicida infection and provide valuable insights into the complex interplay between the host and F. novicida.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"508"},"PeriodicalIF":8.2,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487746/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481419","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
D1-like dopamine receptors promote B-cell differentiation in systemic lupus erythematosus. D1 样多巴胺受体促进系统性红斑狼疮的 B 细胞分化
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-17 DOI: 10.1186/s12964-024-01885-3
Zhongyuan Xiang, Fengxi Wu, Zhenghao He, Fen Tan, Haoran Hu, Chun Zou, Ping Yi, Wenen Liu, Ming Yang

Background: Systemic lupus erythematosus (SLE) is an autoimmune disease that currently cannot be completely cured with a great health burden. Since the production of autoantibodies plays a key role in the pathogenesis of SLE, discovering the underlying immunoregulation mechanism of B cells will be helpful for developing promising immunotherapy for SLE. In recent studies, dopamine receptors (DRDs), G protein-coupled receptors that include D1-like and D2-like subtypes, are expressed on B cells and participate in various physiological processes, involving immune responses. However, the regulatory effect of DRDs on B cells has not been determined.

Methods: This study explored the expression of DRDs on B-cell subsets from SLE patients and healthy individuals. The effects of D1-like receptor on B-cell activation and differentiation were further explored using D1-like receptor agonists or antagonists. RNA-seq and bioinformatics analyses were used to identify specific molecular mechanisms involved.

Results: The D1-like DRDs on B cells of SLE patients were highly expressed compared with those of healthy controls (HCs). D1-like receptor agonist treatment exacerbated lupus-like symptoms in pristane-induced lupus-like mice, while D1-like receptor antagonists alleviated the lupus-like phenotypes. Inhibition of D1-like receptor signals impeded B-cell differentiation, while activation of D1-like receptor signals could promote B cell differentiation. Further RNA-seq confirmed that PTGS2, a gene related to B-cell differentiation, was up-regulated once the D1-like receptor signals were activated, while BMP6 and IL-24 were up-regulated once the D1-like receptor signals were inhibited.

Conclusion: D1-like receptors probably promote B-cell differentiation through the PTGS2/PRDM1 pathway.

背景:系统性红斑狼疮(SLE系统性红斑狼疮(SLE)是一种自身免疫性疾病,目前还不能完全治愈,给患者带来了巨大的健康负担。由于自身抗体的产生在系统性红斑狼疮的发病机制中起着关键作用,因此发现 B 细胞的潜在免疫调节机制将有助于开发治疗系统性红斑狼疮的免疫疗法。近年来的研究发现,多巴胺受体(DRDs)是一种G蛋白偶联受体,包括D1样和D2样亚型,在B细胞上表达,参与各种生理过程,包括免疫反应。然而,DRDs 对 B 细胞的调节作用尚未确定:本研究探讨了 DRDs 在系统性红斑狼疮患者和健康人 B 细胞亚群中的表达。使用D1样受体激动剂或拮抗剂进一步探讨了D1样受体对B细胞活化和分化的影响。通过RNA-seq和生物信息学分析,确定了其中涉及的特定分子机制:结果:与健康对照组(HCs)相比,系统性红斑狼疮患者 B 细胞上的 D1 样 DRDs 高表达。D1样受体激动剂治疗会加重pristane诱导的红斑狼疮样小鼠的红斑狼疮样症状,而D1样受体拮抗剂则会减轻红斑狼疮样表型。抑制D1样受体信号会阻碍B细胞分化,而激活D1样受体信号则能促进B细胞分化。进一步的RNA-seq研究证实,一旦D1样受体信号被激活,与B细胞分化相关的基因PTGS2就会上调,而一旦D1样受体信号被抑制,BMP6和IL-24就会上调:结论:D1样受体可能通过PTGS2/PRDM1途径促进B细胞分化。
{"title":"D1-like dopamine receptors promote B-cell differentiation in systemic lupus erythematosus.","authors":"Zhongyuan Xiang, Fengxi Wu, Zhenghao He, Fen Tan, Haoran Hu, Chun Zou, Ping Yi, Wenen Liu, Ming Yang","doi":"10.1186/s12964-024-01885-3","DOIUrl":"https://doi.org/10.1186/s12964-024-01885-3","url":null,"abstract":"<p><strong>Background: </strong>Systemic lupus erythematosus (SLE) is an autoimmune disease that currently cannot be completely cured with a great health burden. Since the production of autoantibodies plays a key role in the pathogenesis of SLE, discovering the underlying immunoregulation mechanism of B cells will be helpful for developing promising immunotherapy for SLE. In recent studies, dopamine receptors (DRDs), G protein-coupled receptors that include D1-like and D2-like subtypes, are expressed on B cells and participate in various physiological processes, involving immune responses. However, the regulatory effect of DRDs on B cells has not been determined.</p><p><strong>Methods: </strong>This study explored the expression of DRDs on B-cell subsets from SLE patients and healthy individuals. The effects of D1-like receptor on B-cell activation and differentiation were further explored using D1-like receptor agonists or antagonists. RNA-seq and bioinformatics analyses were used to identify specific molecular mechanisms involved.</p><p><strong>Results: </strong>The D1-like DRDs on B cells of SLE patients were highly expressed compared with those of healthy controls (HCs). D1-like receptor agonist treatment exacerbated lupus-like symptoms in pristane-induced lupus-like mice, while D1-like receptor antagonists alleviated the lupus-like phenotypes. Inhibition of D1-like receptor signals impeded B-cell differentiation, while activation of D1-like receptor signals could promote B cell differentiation. Further RNA-seq confirmed that PTGS2, a gene related to B-cell differentiation, was up-regulated once the D1-like receptor signals were activated, while BMP6 and IL-24 were up-regulated once the D1-like receptor signals were inhibited.</p><p><strong>Conclusion: </strong>D1-like receptors probably promote B-cell differentiation through the PTGS2/PRDM1 pathway.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"502"},"PeriodicalIF":8.2,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484144/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481412","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
期刊
Cell Communication and Signaling
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