Melanoma, recognized as the most aggressive type of skin cancer, has experienced a notable increase in cases, especially within populations with fair skin. This highly aggressive cancer is largely driven by UV radiation exposure, resulting in the uncontrolled growth and malignant transformation of melanocytes. The cGAS-STING pathway, an immune signaling mechanism responsible for detecting double-stranded DNA in the cytoplasm, is essential for mediating the immune response against melanoma. This pathway serves a dual purpose: it enhances antitumor immunity by activating immune cells, but it can also promote tumor growth when chronically activated by creating an immunosuppressive environment. This review comprehensively examines the multifaceted implication of the cGAS-STING pathway in melanoma pathogenesis and treatment. We explore its molecular mechanisms, including epigenetic regulation, interaction with signaling pathways such as AR signaling, and modulation by various cellular effectors like TG2 and activin-A. The therapeutic potential of modulating the cGAS-STING pathway is highlighted, with promising results from STING agonists, combination therapies with immune checkpoint inhibitors, and novel drug delivery systems, including nanoparticles and synthetic drugs. Our findings underscore the importance of the cGAS-STING pathway in melanoma, presenting it as a critical target for enhancing anti-tumor immunity. By leveraging this pathway, future therapeutic strategies can potentially convert 'cold' tumors into 'hot' tumors, making them more susceptible to immune responses.
{"title":"cGAS/STING in skin melanoma: from molecular mechanisms to therapeutics.","authors":"Jafaridarabjerdi Mahin, Xuezhu Xu, Ling Li, Cong Zhang","doi":"10.1186/s12964-024-01860-y","DOIUrl":"https://doi.org/10.1186/s12964-024-01860-y","url":null,"abstract":"<p><p>Melanoma, recognized as the most aggressive type of skin cancer, has experienced a notable increase in cases, especially within populations with fair skin. This highly aggressive cancer is largely driven by UV radiation exposure, resulting in the uncontrolled growth and malignant transformation of melanocytes. The cGAS-STING pathway, an immune signaling mechanism responsible for detecting double-stranded DNA in the cytoplasm, is essential for mediating the immune response against melanoma. This pathway serves a dual purpose: it enhances antitumor immunity by activating immune cells, but it can also promote tumor growth when chronically activated by creating an immunosuppressive environment. This review comprehensively examines the multifaceted implication of the cGAS-STING pathway in melanoma pathogenesis and treatment. We explore its molecular mechanisms, including epigenetic regulation, interaction with signaling pathways such as AR signaling, and modulation by various cellular effectors like TG2 and activin-A. The therapeutic potential of modulating the cGAS-STING pathway is highlighted, with promising results from STING agonists, combination therapies with immune checkpoint inhibitors, and novel drug delivery systems, including nanoparticles and synthetic drugs. Our findings underscore the importance of the cGAS-STING pathway in melanoma, presenting it as a critical target for enhancing anti-tumor immunity. By leveraging this pathway, future therapeutic strategies can potentially convert 'cold' tumors into 'hot' tumors, making them more susceptible to immune responses.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"553"},"PeriodicalIF":8.2,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142669854","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}
Background: Postoperative cognitive dysfunction (POCD) is a prevalent complication following anesthesia and surgery, particularly in the elderly, leading to increased mortality and reduced quality of life. Despite its prevalence, there are no effective clinical treatments. Exercise has shown cognitive benefits in aging and various diseases, which can be transferred to sedentary animals through plasma. However, it is unclear if exercise-conditioned plasma can replicate these benefits in the context of POCD.
Methods: Sixteen-month-old male C57BL/6J mice underwent 30 days of voluntary running wheel training or received systemic administration of exercise-conditioned plasma, followed by tibial fracture surgery under general anesthesia at 17 months of age. Cognitive performance, hippocampal synaptic deficits, neuroinflammation, BDNF/TrkB signaling, and medial septum (MS)-hippocampal cholinergic activity were evaluated through immunohistochemical staining, transmission electron microscopy, Western blotting, and biochemical assays. To investigate the role of hippocampal BDNF signaling and cholinergic activity in the therapeutic effects, the TrkB antagonist ANA-12 and the cholinergic receptor muscarinic 1 (CHRM1) antagonist trihexyphenidyl (THP) were administered via intraperitoneal injection, and adeno-associated virus (AAV) vectors expressing Chrm1 shRNA were delivered via intrahippocampal stereotaxic microinjection.
Results: Exercise-conditioned plasma mimicked the benefits of exercise, alleviating cognitive decline induced by anesthesia/surgery, restoring hippocampal synapse formation and levels of regulators for synaptic plasticity, inhibiting neuroinflammatory responses to surgery by microglia and astrocytes, augmenting BDNF production and TrkB phosphorylation in hippocampal neurons, astrocytes, and microglia, upregulating MS expression of choline acetyltransferase (CHAT) and hippocampal expression of CHRM1 in neurons and astrocytes, and enhancing hippocampal cholinergic innervation and acetylcholine release. Conversely, ANA-12 administration blocked TrkB activation and reduced the protective effects on cognition, synaptic deficits, and neuroinflammatory reactivity of glial cells post-surgery. Similarly, THP administration or intrahippocampal delivery of AAV-Chrm1 shRNA inhibited the activation of the hippocampal cholinergic circuit by exercise plasma, negating the cognitive and neuropathological benefits and reducing BDNF/TrkB signaling enhancements.
Conclusion: Exercise-conditioned plasma can replicate the protective effects of exercise against anesthesia/surgery-induced neuroinflammation, synaptic, and cognitive impairments, at least partly, through CHRM1-dependent regulation of hippocampal cholinergic activity and BDNF/TrkB signaling.
{"title":"Exercise-conditioned plasma ameliorates postoperative cognitive dysfunction by activating hippocampal cholinergic circuit and enhancing BDNF/TrkB signaling.","authors":"Xiaodi Lu, Weijie Xiong, Zhuo Chen, Yurou Li, Fengyan Xu, Xue Yang, Meiwen Long, Wenhan Guo, Shuliang Wu, Liang Sun, Guonian Wang","doi":"10.1186/s12964-024-01938-7","DOIUrl":"https://doi.org/10.1186/s12964-024-01938-7","url":null,"abstract":"<p><strong>Background: </strong>Postoperative cognitive dysfunction (POCD) is a prevalent complication following anesthesia and surgery, particularly in the elderly, leading to increased mortality and reduced quality of life. Despite its prevalence, there are no effective clinical treatments. Exercise has shown cognitive benefits in aging and various diseases, which can be transferred to sedentary animals through plasma. However, it is unclear if exercise-conditioned plasma can replicate these benefits in the context of POCD.</p><p><strong>Methods: </strong>Sixteen-month-old male C57BL/6J mice underwent 30 days of voluntary running wheel training or received systemic administration of exercise-conditioned plasma, followed by tibial fracture surgery under general anesthesia at 17 months of age. Cognitive performance, hippocampal synaptic deficits, neuroinflammation, BDNF/TrkB signaling, and medial septum (MS)-hippocampal cholinergic activity were evaluated through immunohistochemical staining, transmission electron microscopy, Western blotting, and biochemical assays. To investigate the role of hippocampal BDNF signaling and cholinergic activity in the therapeutic effects, the TrkB antagonist ANA-12 and the cholinergic receptor muscarinic 1 (CHRM1) antagonist trihexyphenidyl (THP) were administered via intraperitoneal injection, and adeno-associated virus (AAV) vectors expressing Chrm1 shRNA were delivered via intrahippocampal stereotaxic microinjection.</p><p><strong>Results: </strong>Exercise-conditioned plasma mimicked the benefits of exercise, alleviating cognitive decline induced by anesthesia/surgery, restoring hippocampal synapse formation and levels of regulators for synaptic plasticity, inhibiting neuroinflammatory responses to surgery by microglia and astrocytes, augmenting BDNF production and TrkB phosphorylation in hippocampal neurons, astrocytes, and microglia, upregulating MS expression of choline acetyltransferase (CHAT) and hippocampal expression of CHRM1 in neurons and astrocytes, and enhancing hippocampal cholinergic innervation and acetylcholine release. Conversely, ANA-12 administration blocked TrkB activation and reduced the protective effects on cognition, synaptic deficits, and neuroinflammatory reactivity of glial cells post-surgery. Similarly, THP administration or intrahippocampal delivery of AAV-Chrm1 shRNA inhibited the activation of the hippocampal cholinergic circuit by exercise plasma, negating the cognitive and neuropathological benefits and reducing BDNF/TrkB signaling enhancements.</p><p><strong>Conclusion: </strong>Exercise-conditioned plasma can replicate the protective effects of exercise against anesthesia/surgery-induced neuroinflammation, synaptic, and cognitive impairments, at least partly, through CHRM1-dependent regulation of hippocampal cholinergic activity and BDNF/TrkB signaling.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"551"},"PeriodicalIF":8.2,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142669865","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}
Pub Date : 2024-11-18DOI: 10.1186/s12964-024-01936-9
Iulia Andreea Pelisenco, Daniela Zizioli, Flora Guerra, Ilaria Grossi, Cecilia Bucci, Luca Mignani, Giulia Girolimetti, Riccardo Di Corato, Vito Giuseppe D'Agostino, Eleonora Marchina, Giuseppina De Petro, Alessandro Salvi
Background: Extracellular vesicles (EVs) are a group of nanoscale cell-derived membranous structures secreted by all cell types, containing molecular cargoes involved in intercellular communication. EVs can be used to mimic "nature's delivery system" to transport nucleic acids, peptides, lipids, and metabolites to target recipient cells. EVs offer a range of advantages over traditional synthetic carriers, thus paving the way for innovative drug delivery approaches that can be used in different diseases, including cancer. Here, by using breast cancer (BC) cells treated with the multi-kinase inhibitor sorafenib, we generated EVs enriched in specific non-coding RNAs (miR-23b-3p, miR-126-3p, and the long ncRNA GAS5) and investigated their potential impact on the aggressive properties of the BC in vitro and in vivo using zebrafish.
Methods: EVs were collected from 4 different BC cell lines (HCC1937, MDA-MB-231, MCF-7, and MDA-MB-453) and characterized by western blotting, transmission electron microscopy and nanoparticle tracking analysis. Levels of encapsulated miR-23b-3p, miR-126-3p, and GAS5 were quantified by ddPCR. The role of the EVs as carriers of ncRNAs in vivo was established by injecting MDA-MB-231 and MDA-MB-453 cells into zebrafish embryos followed by EV-based treatment of the xenografts with EVs rich in miR-23b-3p, miR-126-3p and GAS5.
Results: ddPCR analysis revealed elevated levels of miR-23b-3p, miR-126-3p, and GAS5, encapsulated in the EVs released by the aforementioned cell lines, following sorafenib treatment. The use of EVs as carriers of these specific ncRNAs in the treatment of BC cells resulted in a significant increase in the expression levels of the three ncRNAs along with the inhibition of cellular proliferation in vitro. In vivo experiments demonstrated a remarkable reduction of xenograft tumor area, suppression of angiogenesis, and decreased number of micrometastasis in the tails after administration of EVs enriched with these ncRNAs.
Conclusions: Our study demonstrated that sorafenib-induced EVs, enriched with specific tumor-suppressor ncRNAs, can effectively inhibit the aggressive BC characteristics in vitro and in vivo. Our findings indicate an alternative way to enrich EVs with specific tumor-suppressor ncRNAs by treating the cells with an anticancer drug and support the development of new potential experimental molecular approaches to target the aggressive properties of cancer cells.
背景:细胞外囊泡(EVs)是一类由所有类型细胞分泌的纳米级细胞衍生膜结构,内含参与细胞间通讯的分子货物。EVs可用于模拟 "大自然的输送系统",将核酸、肽、脂质和代谢物输送到目标受体细胞。与传统的人工合成载体相比,EVs 具有一系列优势,从而为可用于不同疾病(包括癌症)的创新药物递送方法铺平了道路。在这里,我们利用接受多激酶抑制剂索拉非尼治疗的乳腺癌(BC)细胞,生成了富含特定非编码RNA(miR-23b-3p、miR-126-3p和长ncRNA GAS5)的EVs,并利用斑马鱼研究了它们对体外和体内BC侵袭性的潜在影响:从4种不同的BC细胞系(HCC1937、MDA-MB-231、MCF-7和MDA-MB-453)中收集EVs,并通过Western印迹、透射电子显微镜和纳米粒子追踪分析对其进行表征。通过 ddPCR 对包裹的 miR-23b-3p、miR-126-3p 和 GAS5 的水平进行了量化。通过将 MDA-MB-231 和 MDA-MB-453 细胞注射到斑马鱼胚胎中,然后用富含 miR-23b-3p、miR-126-3p 和 GAS5 的 EV 处理异种移植,确定了 EV 在体内作为 ncRNAs 载体的作用。结果:ddPCR 分析显示,索拉非尼处理后,上述细胞系释放的 EV 中包裹的 miR-23b-3p、miR-126-3p 和 GAS5 水平升高。用 EVs 作为这些特定 ncRNAs 的载体处理 BC 细胞,可显著提高这三种 ncRNAs 的表达水平,同时抑制体外细胞增殖。体内实验表明,服用富含这些ncRNAs的EVs后,异种移植肿瘤面积明显缩小,血管生成受到抑制,尾部微转移灶数量减少:我们的研究表明,富含特定抑瘤ncRNA的索拉非尼诱导的EVs能在体外和体内有效抑制侵袭性BC特征。我们的研究结果表明,用抗癌药物处理细胞是富集具有特异性肿瘤抑制ncRNAs的EVs的另一种方法,并支持开发新的针对癌细胞侵袭性的潜在实验分子方法。
{"title":"miR-23b-3p, miR-126-3p and GAS5 delivered by extracellular vesicles inhibit breast cancer xenografts in zebrafish.","authors":"Iulia Andreea Pelisenco, Daniela Zizioli, Flora Guerra, Ilaria Grossi, Cecilia Bucci, Luca Mignani, Giulia Girolimetti, Riccardo Di Corato, Vito Giuseppe D'Agostino, Eleonora Marchina, Giuseppina De Petro, Alessandro Salvi","doi":"10.1186/s12964-024-01936-9","DOIUrl":"https://doi.org/10.1186/s12964-024-01936-9","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs) are a group of nanoscale cell-derived membranous structures secreted by all cell types, containing molecular cargoes involved in intercellular communication. EVs can be used to mimic \"nature's delivery system\" to transport nucleic acids, peptides, lipids, and metabolites to target recipient cells. EVs offer a range of advantages over traditional synthetic carriers, thus paving the way for innovative drug delivery approaches that can be used in different diseases, including cancer. Here, by using breast cancer (BC) cells treated with the multi-kinase inhibitor sorafenib, we generated EVs enriched in specific non-coding RNAs (miR-23b-3p, miR-126-3p, and the long ncRNA GAS5) and investigated their potential impact on the aggressive properties of the BC in vitro and in vivo using zebrafish.</p><p><strong>Methods: </strong>EVs were collected from 4 different BC cell lines (HCC1937, MDA-MB-231, MCF-7, and MDA-MB-453) and characterized by western blotting, transmission electron microscopy and nanoparticle tracking analysis. Levels of encapsulated miR-23b-3p, miR-126-3p, and GAS5 were quantified by ddPCR. The role of the EVs as carriers of ncRNAs in vivo was established by injecting MDA-MB-231 and MDA-MB-453 cells into zebrafish embryos followed by EV-based treatment of the xenografts with EVs rich in miR-23b-3p, miR-126-3p and GAS5.</p><p><strong>Results: </strong>ddPCR analysis revealed elevated levels of miR-23b-3p, miR-126-3p, and GAS5, encapsulated in the EVs released by the aforementioned cell lines, following sorafenib treatment. The use of EVs as carriers of these specific ncRNAs in the treatment of BC cells resulted in a significant increase in the expression levels of the three ncRNAs along with the inhibition of cellular proliferation in vitro. In vivo experiments demonstrated a remarkable reduction of xenograft tumor area, suppression of angiogenesis, and decreased number of micrometastasis in the tails after administration of EVs enriched with these ncRNAs.</p><p><strong>Conclusions: </strong>Our study demonstrated that sorafenib-induced EVs, enriched with specific tumor-suppressor ncRNAs, can effectively inhibit the aggressive BC characteristics in vitro and in vivo. Our findings indicate an alternative way to enrich EVs with specific tumor-suppressor ncRNAs by treating the cells with an anticancer drug and support the development of new potential experimental molecular approaches to target the aggressive properties of cancer cells.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"552"},"PeriodicalIF":8.2,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142669870","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}
Pub Date : 2024-11-18DOI: 10.1186/s12964-024-01926-x
Yongkang Xu, Jiayu Zeng, Kan Liu, Dan Li, Shenglan Huang, Shumin Fu, Mao Ye, Si Tao, Jianbing Wu
Background: The relationship between hepatocellular carcinoma (HCC) metastasis and cancer metabolism reprogramming is becoming increasingly evident. Ubiquitin-specific protease 11 (USP11), a member of the deubiquitinating enzyme family, has been linked to various cancer-related processes. While USP11 is known to promote HCC metastasis and proliferation, the precise mechanisms, especially those related to cancer metabolism, remain unclear.
Methods: Through mass spectrometry, co-immunoprecipitation, immunofluorescence, and ubiquitination assays, we identified USP11 as the key deubiquitinase for SREBF1.Lipogenesis was evaluated using Oil Red O and Nile Red staining, along with the detection of triglycerides and cholesterol. To assess HCC cell proliferation, migration, and invasion in vitro, Transwell assays, EDU, colony formation, and CCK-8 were conducted. Xenograft models in nude mice were developed to verify the role of the USP11/SREBF1 axis in lipogenesis and tumor growth in vivo.
Results: USP11 directly interacts with SREBF1, and its silencing leads to the disruption of SREBF1 stabilization through K48-linked deubiquitination and degradation. Importantly, the truncated mutant USP11 (503-938 aa) interacts with the truncated mutant SREBF1 (569-1147aa), with K1151 playing a crucial role in this interaction. Higher levels of USP11 enhance lipogenesis, proliferation, and metastasis in HCC cells. Importantly, the knockdown of SREBF1 weakened the effects of USP11 in enhancing lipogenesis and tumorigenesis. Futhermore, the elevated expression of USP11 and SREBF1 in HCC tissue serves as an indicator of poor prognosis in HCC patients.
Conclusions: In summary, our study reveals that USP11 promotes HCC proliferation and metastasis through SREBF1-induced lipogenesis. These findings provide a foundation for novel therapies targeting lipid metabolism in HCC.
{"title":"USP11 promotes lipogenesis and tumorigenesis by regulating SREBF1 stability in hepatocellular carcinoma.","authors":"Yongkang Xu, Jiayu Zeng, Kan Liu, Dan Li, Shenglan Huang, Shumin Fu, Mao Ye, Si Tao, Jianbing Wu","doi":"10.1186/s12964-024-01926-x","DOIUrl":"https://doi.org/10.1186/s12964-024-01926-x","url":null,"abstract":"<p><strong>Background: </strong>The relationship between hepatocellular carcinoma (HCC) metastasis and cancer metabolism reprogramming is becoming increasingly evident. Ubiquitin-specific protease 11 (USP11), a member of the deubiquitinating enzyme family, has been linked to various cancer-related processes. While USP11 is known to promote HCC metastasis and proliferation, the precise mechanisms, especially those related to cancer metabolism, remain unclear.</p><p><strong>Methods: </strong>Through mass spectrometry, co-immunoprecipitation, immunofluorescence, and ubiquitination assays, we identified USP11 as the key deubiquitinase for SREBF1.Lipogenesis was evaluated using Oil Red O and Nile Red staining, along with the detection of triglycerides and cholesterol. To assess HCC cell proliferation, migration, and invasion in vitro, Transwell assays, EDU, colony formation, and CCK-8 were conducted. Xenograft models in nude mice were developed to verify the role of the USP11/SREBF1 axis in lipogenesis and tumor growth in vivo.</p><p><strong>Results: </strong>USP11 directly interacts with SREBF1, and its silencing leads to the disruption of SREBF1 stabilization through K48-linked deubiquitination and degradation. Importantly, the truncated mutant USP11 (503-938 aa) interacts with the truncated mutant SREBF1 (569-1147aa), with K1151 playing a crucial role in this interaction. Higher levels of USP11 enhance lipogenesis, proliferation, and metastasis in HCC cells. Importantly, the knockdown of SREBF1 weakened the effects of USP11 in enhancing lipogenesis and tumorigenesis. Futhermore, the elevated expression of USP11 and SREBF1 in HCC tissue serves as an indicator of poor prognosis in HCC patients.</p><p><strong>Conclusions: </strong>In summary, our study reveals that USP11 promotes HCC proliferation and metastasis through SREBF1-induced lipogenesis. These findings provide a foundation for novel therapies targeting lipid metabolism in HCC.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"550"},"PeriodicalIF":8.2,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142669873","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}
Pub Date : 2024-11-16DOI: 10.1186/s12964-024-01930-1
Duško Lainšček, Simon Horvat, Klemen Dolinar, Filip Ivanovski, Rok Romih, Sergej Pirkmajer, Roman Jerala, Mateja Manček-Keber
Various signaling pathways are essential for both the innate immune response and the maintenance of cell homeostasis, requiring coordinated interactions among them. In this study, a mutation in the caspase-1 recognition site within MyD88 abolished inflammasome-dependent negative regulation, causing phenotypic changes in mice with some similarities to human NEMO-deficiencies. The MyD88D162E mutation reduced MyD88 protein levels and colon inflammation in DSS-induced colitis mice but did not affect cytokine expression in bone marrow-derived macrophages (BMDMs). However, compared to MyD88wt counterparts, MyD88D162E BMDMs had increased oxidative stress and dysfunctional mitochondria, along with reduced prosurvival Bcl-xL and BTK expression, rendering cells more prone to apoptosis, exacerbated by ibrutinib treatment. NF-κB activation by lipopolysaccharide mitigated this sensitive phenotype. These findings underscore the importance of MyD88wt signaling for NF-κB activation, protecting against macrophage premature apoptosis at resting state. Targeting MyD88 quantity rather than just its signaling could be a promising strategy for MyD88-driven lymphoma treatment.
{"title":"MyD88 protein destabilization mitigates NF-κB-dependent protection against macrophage apoptosis.","authors":"Duško Lainšček, Simon Horvat, Klemen Dolinar, Filip Ivanovski, Rok Romih, Sergej Pirkmajer, Roman Jerala, Mateja Manček-Keber","doi":"10.1186/s12964-024-01930-1","DOIUrl":"10.1186/s12964-024-01930-1","url":null,"abstract":"<p><p>Various signaling pathways are essential for both the innate immune response and the maintenance of cell homeostasis, requiring coordinated interactions among them. In this study, a mutation in the caspase-1 recognition site within MyD88 abolished inflammasome-dependent negative regulation, causing phenotypic changes in mice with some similarities to human NEMO-deficiencies. The MyD88<sup>D162E</sup> mutation reduced MyD88 protein levels and colon inflammation in DSS-induced colitis mice but did not affect cytokine expression in bone marrow-derived macrophages (BMDMs). However, compared to MyD88<sup>wt</sup> counterparts, MyD88<sup>D162E</sup> BMDMs had increased oxidative stress and dysfunctional mitochondria, along with reduced prosurvival Bcl-xL and BTK expression, rendering cells more prone to apoptosis, exacerbated by ibrutinib treatment. NF-κB activation by lipopolysaccharide mitigated this sensitive phenotype. These findings underscore the importance of MyD88<sup>wt</sup> signaling for NF-κB activation, protecting against macrophage premature apoptosis at resting state. Targeting MyD88 quantity rather than just its signaling could be a promising strategy for MyD88-driven lymphoma treatment.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"549"},"PeriodicalIF":8.2,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11568545/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142645236","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}
Pub Date : 2024-11-15DOI: 10.1186/s12964-024-01932-z
Zixi Liu, Simiao Xu, Lin Chen, Jun Gong, Min Wang
Pyroptosis is a lytic and inflammatory form of gasdermin protein-mediated programmed cell death that is typically initiated by inflammasomes. The inflammasome response is an effective mechanism for eradicating germs and cancer cells in the event of cellular injury. The gasdermin family is responsible for initiating pyroptosis, a process in which holes are made in the cell membrane to allow inflammatory chemicals to escape. Mounting evidence indicates that pyroptosis is critical for controlling the development of cancer. In this review, we provide a general overview of pyroptosis, examine the relationship between the primary elements of pyroptosis and tumors, and stress the necessity of pyroptosis-targeted therapy in tumors. Furthermore, we explore its dual nature as a double-edged sword capable of both inhibiting and facilitating the growth of cancer, depending on the specific conditions. Ultimately, pyroptosis is a phenomenon that has both positive and negative effects on tumors. Using this dual impact in a reasonable manner may facilitate investigation into the initiation and progression of tumors and offer insights for the development of novel treatments centered on pyroptosis.
{"title":"The role of pyroptosis in cancer: key components and therapeutic potential.","authors":"Zixi Liu, Simiao Xu, Lin Chen, Jun Gong, Min Wang","doi":"10.1186/s12964-024-01932-z","DOIUrl":"10.1186/s12964-024-01932-z","url":null,"abstract":"<p><p>Pyroptosis is a lytic and inflammatory form of gasdermin protein-mediated programmed cell death that is typically initiated by inflammasomes. The inflammasome response is an effective mechanism for eradicating germs and cancer cells in the event of cellular injury. The gasdermin family is responsible for initiating pyroptosis, a process in which holes are made in the cell membrane to allow inflammatory chemicals to escape. Mounting evidence indicates that pyroptosis is critical for controlling the development of cancer. In this review, we provide a general overview of pyroptosis, examine the relationship between the primary elements of pyroptosis and tumors, and stress the necessity of pyroptosis-targeted therapy in tumors. Furthermore, we explore its dual nature as a double-edged sword capable of both inhibiting and facilitating the growth of cancer, depending on the specific conditions. Ultimately, pyroptosis is a phenomenon that has both positive and negative effects on tumors. Using this dual impact in a reasonable manner may facilitate investigation into the initiation and progression of tumors and offer insights for the development of novel treatments centered on pyroptosis.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"548"},"PeriodicalIF":8.2,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11566483/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142645240","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}
Background: Polyploid giant cancer cells (PGCCs) have properties of cancer stem cells (CSCs). PGCCs with daughter cells (PDCs) undergo epithelial-mesenchymal transition and show enhanced cellular plasticity. This study aimed to elucidate the mechanisms underlying the osteo/chondrogenic-like differentiation of PDCs, which may be exploited therapeutically by transdifferentiation into post-mitotic and functional cells.
Methods: Cobalt chloride was used to induce PGCC formation in MDA-MB-231 and HEY cells, and PDCs were cultured in osteo/chondrogenic differentiation media. Alcian blue staining was used to confirm osteo/chondrogenic differentiation, and the cell cycle was detected using flow cytometry. The expression of osteo/chondrogenic differentiation-related proteins was compared, and a co-immunoprecipitation assay was used to demonstrate the interactions between proteins. Bioinformatic analysis was used to explore the regulatory mechanism of osteo/chondrogenic differentiation, and a dual-luciferase reporter assay was performed to validate the interaction between transcriptional factors and target genes. Animal xenograft models were used to confirm the osteo/chondrogenic differentiation of PDCs.
Results: When cultured in osteo/chondrogenic medium, the stemness of PDCs decreased, and the expression of osteo/chondrogenic-related markers increased. This osteo/chondrogenic-like process was regulated by the transforming growth factor-β pathway in a time-dependent manner. A concurrent increase in the expression of histone acetyltransferase p300 and the transcription factor CCCTC-binding factor (CTCF) was observed. Co-immunoprecipitation assays revealed that p300 acetylated the osteo/chondrogenic marker RUNT-related transcription factor 2 (RUNX2). Analysis of chromatin immunoprecipitation sequencing datasets revealed that both CTCF and histone H3 lysine 27 acetylation (H3K27ac) were enriched in the promoter region of E1A-associated protein p300 (P300). The four predicted binding sites for CTCF and P300 were validated using dual-luciferase reporter assays. We examined the interaction between CTCF and H3K27ac and found that these two proteins had a combined effect on the transactivation of P300.
Conclusion: CTCF, in synergy with H3K27ac, amplified the expression of P300, facilitating acetyl group transfer to RUNX2. This acetylation stabilized RUNX2 and promoted osteo/chondrogenic differentiation, thereby reducing the incidence of PDC malignancies.
{"title":"Role of the CTCF/p300 axis in osteochondrogenic-like differentiation of polyploid giant cancer cells with daughter cells.","authors":"Xiaohui Yang, Jie Sun, Yidi Ning, Jiangping Wang, Jing Xu, Shiwu Zhang","doi":"10.1186/s12964-024-01933-y","DOIUrl":"10.1186/s12964-024-01933-y","url":null,"abstract":"<p><strong>Background: </strong>Polyploid giant cancer cells (PGCCs) have properties of cancer stem cells (CSCs). PGCCs with daughter cells (PDCs) undergo epithelial-mesenchymal transition and show enhanced cellular plasticity. This study aimed to elucidate the mechanisms underlying the osteo/chondrogenic-like differentiation of PDCs, which may be exploited therapeutically by transdifferentiation into post-mitotic and functional cells.</p><p><strong>Methods: </strong>Cobalt chloride was used to induce PGCC formation in MDA-MB-231 and HEY cells, and PDCs were cultured in osteo/chondrogenic differentiation media. Alcian blue staining was used to confirm osteo/chondrogenic differentiation, and the cell cycle was detected using flow cytometry. The expression of osteo/chondrogenic differentiation-related proteins was compared, and a co-immunoprecipitation assay was used to demonstrate the interactions between proteins. Bioinformatic analysis was used to explore the regulatory mechanism of osteo/chondrogenic differentiation, and a dual-luciferase reporter assay was performed to validate the interaction between transcriptional factors and target genes. Animal xenograft models were used to confirm the osteo/chondrogenic differentiation of PDCs.</p><p><strong>Results: </strong>When cultured in osteo/chondrogenic medium, the stemness of PDCs decreased, and the expression of osteo/chondrogenic-related markers increased. This osteo/chondrogenic-like process was regulated by the transforming growth factor-β pathway in a time-dependent manner. A concurrent increase in the expression of histone acetyltransferase p300 and the transcription factor CCCTC-binding factor (CTCF) was observed. Co-immunoprecipitation assays revealed that p300 acetylated the osteo/chondrogenic marker RUNT-related transcription factor 2 (RUNX2). Analysis of chromatin immunoprecipitation sequencing datasets revealed that both CTCF and histone H3 lysine 27 acetylation (H3K27ac) were enriched in the promoter region of E1A-associated protein p300 (P300). The four predicted binding sites for CTCF and P300 were validated using dual-luciferase reporter assays. We examined the interaction between CTCF and H3K27ac and found that these two proteins had a combined effect on the transactivation of P300.</p><p><strong>Conclusion: </strong>CTCF, in synergy with H3K27ac, amplified the expression of P300, facilitating acetyl group transfer to RUNX2. This acetylation stabilized RUNX2 and promoted osteo/chondrogenic differentiation, thereby reducing the incidence of PDC malignancies.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"546"},"PeriodicalIF":8.2,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11566548/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142645238","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}
Fusobacterium nucleatum (Fn) has been extensively studied for its connection to colorectal cancer (CRC) and its potential role in chemotherapy resistance. Studies indicate that Fn is commonly found in CRC tissues and is associated with unfavorable prognosis and treatment failure. It has been shown that Fn promotes chemoresistance by affecting autophagy, a cellular process that helps cells survive under stressful conditions. Additionally, Fn targets specific signaling pathways that activate particular microRNAs and modulate the response to chemotherapy. Understanding the current molecular mechanisms and investigating the importance of Fn-inducing chemoresistance could provide valuable insights for developing novel therapies. This review surveys the role of Fn in tumor proliferation, metastasis, and chemoresistance in CRC, focusing on its effects on the tumor microenvironment, gene expression, and resistance to conventional chemotherapy drugs. It also discusses the therapeutic implications of targeting Fn in CRC treatment and highlights the need for further research.
{"title":"Exploring the role of Fusobacterium nucleatum in colorectal cancer: implications for tumor proliferation and chemoresistance.","authors":"Leila Dadgar-Zankbar, Zahra Elahi, Aref Shariati, Azad Khaledi, Shabnam Razavi, Amin Khoshbayan","doi":"10.1186/s12964-024-01909-y","DOIUrl":"10.1186/s12964-024-01909-y","url":null,"abstract":"<p><p>Fusobacterium nucleatum (Fn) has been extensively studied for its connection to colorectal cancer (CRC) and its potential role in chemotherapy resistance. Studies indicate that Fn is commonly found in CRC tissues and is associated with unfavorable prognosis and treatment failure. It has been shown that Fn promotes chemoresistance by affecting autophagy, a cellular process that helps cells survive under stressful conditions. Additionally, Fn targets specific signaling pathways that activate particular microRNAs and modulate the response to chemotherapy. Understanding the current molecular mechanisms and investigating the importance of Fn-inducing chemoresistance could provide valuable insights for developing novel therapies. This review surveys the role of Fn in tumor proliferation, metastasis, and chemoresistance in CRC, focusing on its effects on the tumor microenvironment, gene expression, and resistance to conventional chemotherapy drugs. It also discusses the therapeutic implications of targeting Fn in CRC treatment and highlights the need for further research.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"547"},"PeriodicalIF":8.2,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11566256/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142645234","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}
Background: Endocrine therapy (ET) has improved the clinical outcomes of Estrogen receptor alpha-positive (ERɑ +) breast cancer (BC) patients, even though resistance to ET remains a clinical issue. Mutations in the hormone-binding domain of ERɑ represent an acquired intrinsic mechanism of ET resistance. However, the latter also depends on the multiple functional interactions between BC cells and the tumor microenvironment (TME). Here, we investigated how the most common Y537S-ERɑ mutation may influence the behavior of fibroblasts, the most prominent component of the TME.
Methods: We conducted coculture experiments with normal human foreskin fibroblasts BJ1-hTERT (NFs), cancer-associated fibroblasts (CAFs), isolated from human BC specimens, and Y537S CRISPR-expressing MCF-7 BC cells (MCF-7YS). Mass spectrometry (MS) and Metacore analyses were performed to investigate how the functional interactions between BC cells/fibroblasts may affect their proteomic profile. The impact of fibroblasts on BC tumor growth and metastatic potential was evaluated in nude mice.
Results: Mutant BC conditioned medium (CM) affected the morphology/proliferation/migration of both NFs and CAFs. 198 deregulated proteins signed the proteomic similarity profile of NFs exposed to the YS-CM and CAFs. Among the upregulated proteins, Yes-associated protein 1 (YAP1) was the main central hub in the direct interaction network. Increased YAP1 protein expression and activity were confirmed in NFs treated with MCF-7YS-CM. However, YAP1 activation appears to crosstalk with the insulin growth factor-1 receptor (IGF-1R). Higher amount of IGF-1 were noticed in the MCF-7YS-CM cells compared to the MCF-7P, and IGF-1 immunodepletion reversed the enhanced YAP1 expression and activity. Mutant cells upon exposure to the NF- and CAF-CM exhibited an enhanced proliferation/growth/migration/invasion compared to the MCF-7P. MCF-7YS cells when implanted with CAFs showed an early relative increased tumor volume compared to YS alone. No changes were observed when MCF-7P cells were co-implanted with CAFs. Compared with that in MCF-7P cells, the metastatic burden of MCF-7YS cells was intrinsically greater, and this effect was augmented upon treatment with NF-CM and further increased with CAF-CM.
Conclusions: YS mutant BC cells induced the conversion of fibroblasts into CAFs, via YAP, which represent a potential therapeutic target which interrupt the functional interactions between mutant cells/TME and to be implemented in the novel therapeutic strategy of a subset of metastatic BC patients carrying the frequent Y537S mutations.
{"title":"Evidence that CRISPR-Cas9 Y537S-mutant expressing breast cancer cells activate Yes-associated protein 1 to driving the conversion of normal fibroblasts into cancer-associated fibroblasts.","authors":"Luca Gelsomino, Amanda Caruso, Emine Tasan, Adele Elisabetta Leonetti, Rocco Malivindi, Giuseppina Daniela Naimo, Francesca Giordano, Salvatore Panza, Guowei Gu, Benedetta Perrone, Cinzia Giordano, Loredana Mauro, Bruno Nardo, Gianfranco Filippelli, Daniela Bonofiglio, Ines Barone, Suzanne A W Fuqua, Stefania Catalano, Sebastiano Andò","doi":"10.1186/s12964-024-01918-x","DOIUrl":"10.1186/s12964-024-01918-x","url":null,"abstract":"<p><strong>Background: </strong>Endocrine therapy (ET) has improved the clinical outcomes of Estrogen receptor alpha-positive (ERɑ +) breast cancer (BC) patients, even though resistance to ET remains a clinical issue. Mutations in the hormone-binding domain of ERɑ represent an acquired intrinsic mechanism of ET resistance. However, the latter also depends on the multiple functional interactions between BC cells and the tumor microenvironment (TME). Here, we investigated how the most common Y537S-ERɑ mutation may influence the behavior of fibroblasts, the most prominent component of the TME.</p><p><strong>Methods: </strong>We conducted coculture experiments with normal human foreskin fibroblasts BJ1-hTERT (NFs), cancer-associated fibroblasts (CAFs), isolated from human BC specimens, and Y537S CRISPR-expressing MCF-7 BC cells (MCF-7YS). Mass spectrometry (MS) and Metacore analyses were performed to investigate how the functional interactions between BC cells/fibroblasts may affect their proteomic profile. The impact of fibroblasts on BC tumor growth and metastatic potential was evaluated in nude mice.</p><p><strong>Results: </strong>Mutant BC conditioned medium (CM) affected the morphology/proliferation/migration of both NFs and CAFs. 198 deregulated proteins signed the proteomic similarity profile of NFs exposed to the YS-CM and CAFs. Among the upregulated proteins, Yes-associated protein 1 (YAP1) was the main central hub in the direct interaction network. Increased YAP1 protein expression and activity were confirmed in NFs treated with MCF-7YS-CM. However, YAP1 activation appears to crosstalk with the insulin growth factor-1 receptor (IGF-1R). Higher amount of IGF-1 were noticed in the MCF-7YS-CM cells compared to the MCF-7P, and IGF-1 immunodepletion reversed the enhanced YAP1 expression and activity. Mutant cells upon exposure to the NF- and CAF-CM exhibited an enhanced proliferation/growth/migration/invasion compared to the MCF-7P. MCF-7YS cells when implanted with CAFs showed an early relative increased tumor volume compared to YS alone. No changes were observed when MCF-7P cells were co-implanted with CAFs. Compared with that in MCF-7P cells, the metastatic burden of MCF-7YS cells was intrinsically greater, and this effect was augmented upon treatment with NF-CM and further increased with CAF-CM.</p><p><strong>Conclusions: </strong>YS mutant BC cells induced the conversion of fibroblasts into CAFs, via YAP, which represent a potential therapeutic target which interrupt the functional interactions between mutant cells/TME and to be implemented in the novel therapeutic strategy of a subset of metastatic BC patients carrying the frequent Y537S mutations.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"545"},"PeriodicalIF":8.2,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11566413/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142633233","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}
Pub Date : 2024-11-14DOI: 10.1186/s12964-024-01910-5
Sadiya Bi Shaikh, Rex Devasahayam Arokia Balaya, Shobha Dagamajalu, Yashodhar Prabhakar Bhandary, Hoshang Unwalla, Thottethodi Subrahmanya Keshava Prasad, Irfan Rahman
Plasminogen activator inhibitor-1 (PAI-1) is a vital regulator of the fibrinolytic mechanism and has been intricately involved in various physiological and clinical processes, including cancer, thrombosis, and wound healing. The PAI-1 signaling pathway is multifaceted, encompassing numerous signaling molecules and nodes. Recent studies have revealed a novel contribution of PAI-1 during cellular senescence. This review introduces a pathway resource detailing the signaling network events mediated by PAI-1. The literature curated on the PAI-1 system was manually compiled from various published studies, our analysis presents a signaling pathway network of PAI-1, which includes various events like enzyme catalysis, molecular association, gene regulation, protein expression, and protein translocation. This signaling network aims to provide a detailed analysis of the existing understanding of the PAI-1 signaling pathway in the context of cellular senescence across various research models. By developing this pathway, we aspire to deepen our understanding of aging and senescence research, ultimately contributing to the pursuit of effective therapeutic approaches for these complex chronic diseases.
{"title":"A signaling pathway map of plasminogen activator inhibitor-1 (PAI-1/SERPINE-1): a review of an innovative frontier in molecular aging and cellular senescence.","authors":"Sadiya Bi Shaikh, Rex Devasahayam Arokia Balaya, Shobha Dagamajalu, Yashodhar Prabhakar Bhandary, Hoshang Unwalla, Thottethodi Subrahmanya Keshava Prasad, Irfan Rahman","doi":"10.1186/s12964-024-01910-5","DOIUrl":"10.1186/s12964-024-01910-5","url":null,"abstract":"<p><p>Plasminogen activator inhibitor-1 (PAI-1) is a vital regulator of the fibrinolytic mechanism and has been intricately involved in various physiological and clinical processes, including cancer, thrombosis, and wound healing. The PAI-1 signaling pathway is multifaceted, encompassing numerous signaling molecules and nodes. Recent studies have revealed a novel contribution of PAI-1 during cellular senescence. This review introduces a pathway resource detailing the signaling network events mediated by PAI-1. The literature curated on the PAI-1 system was manually compiled from various published studies, our analysis presents a signaling pathway network of PAI-1, which includes various events like enzyme catalysis, molecular association, gene regulation, protein expression, and protein translocation. This signaling network aims to provide a detailed analysis of the existing understanding of the PAI-1 signaling pathway in the context of cellular senescence across various research models. By developing this pathway, we aspire to deepen our understanding of aging and senescence research, ultimately contributing to the pursuit of effective therapeutic approaches for these complex chronic diseases.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"544"},"PeriodicalIF":8.2,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11566301/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142633196","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}