首页 > 最新文献

Cell Communication and Signaling最新文献

英文 中文
COVID-19 patient serum-derived extracellular vesicles deliver miR-20b-5p induces neutrophil extracellular traps.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1186/s12964-025-02095-1
Yao Liao, Yuheng Liu, Dinghao Li, Shiqi Luo, Yun Huang, Junwei Wu, Jin Su, Yi Yang, Ji Wu, Zifeng Zhu, Mengxi Yanglan, Haiyi Deng, Xinyi Wu, Junhao Xu, Feiyang Cao, Chunmei Cai, Zhen Li, Ruibing Yang, Xiaoyan Deng, Jie Wei, Lifu Wang

Background: Severe cases of COVID-19 are characterized by an excessive presence of neutrophils. Neutrophil extracellular traps (NETs), released by activated neutrophils due to SARS-CoV-2 infection, contribute to lung epithelial cell death and are key drivers in COVID-19-associated immunothrombosis. However, the mechanism underlying NET formation in COVID-19 remain unclear.

Methods: Extracellular vesicles (EVs) were isolated from the serum of COVID-19 patients and healthy volunteers, while neutrophils were isolated from blood samples of healthy volunteers. Neutrophils were treated with EVs, and the formation of NETs was observed. To identify the components responsible for the COVID-19-EVs-induced NET formation, we analyzed the expression profiles of microRNA (miRNAs) in COVID-19-EVs. We identified eight highly expressed miRNAs in COVID-19-EVs and explored their potential roles in COVID-19-EVs-mediated NET formation. Additionally, we explored the role of miR-20b-5p in COVID-19-EVs-induced NET formation.

Results: In this study, we demonstrate that patients with COVID-19 have a higher concentration of serum EVs (COVID-19-EVs) than healthy controls (Normal-EVs). We also found that COVID-19-EVs are internalized by neutrophils to induced NET formation. Through comprehensive miRNA profiling of COVID-19-EVs versus Normal-EVs, we identified 78 differentially expressed miRNAs, with 27 of these being upregulated and 51 being downregulated. Subsequently, we discovered that COVID-19-EVs that were highly abundant with certain miRNAs promote NET formation. Specifically, miR-20b-5p was found to be the strongest inducer of NET formation of the identified miRNAs. Inhibition of miR-20b-5p resulted in a significant decrease in COVID-19-EVs-mediated induction of NET formation.

Conclusion: Herein, we reveal a previously unknown role of COVID-19-EVs in NET formation, which contributes to COVID-19 progression. This study suggests that miR-20b-5p may serve as a potential therapeutic target for COVID-19 treatment.

{"title":"COVID-19 patient serum-derived extracellular vesicles deliver miR-20b-5p induces neutrophil extracellular traps.","authors":"Yao Liao, Yuheng Liu, Dinghao Li, Shiqi Luo, Yun Huang, Junwei Wu, Jin Su, Yi Yang, Ji Wu, Zifeng Zhu, Mengxi Yanglan, Haiyi Deng, Xinyi Wu, Junhao Xu, Feiyang Cao, Chunmei Cai, Zhen Li, Ruibing Yang, Xiaoyan Deng, Jie Wei, Lifu Wang","doi":"10.1186/s12964-025-02095-1","DOIUrl":"https://doi.org/10.1186/s12964-025-02095-1","url":null,"abstract":"<p><strong>Background: </strong>Severe cases of COVID-19 are characterized by an excessive presence of neutrophils. Neutrophil extracellular traps (NETs), released by activated neutrophils due to SARS-CoV-2 infection, contribute to lung epithelial cell death and are key drivers in COVID-19-associated immunothrombosis. However, the mechanism underlying NET formation in COVID-19 remain unclear.</p><p><strong>Methods: </strong>Extracellular vesicles (EVs) were isolated from the serum of COVID-19 patients and healthy volunteers, while neutrophils were isolated from blood samples of healthy volunteers. Neutrophils were treated with EVs, and the formation of NETs was observed. To identify the components responsible for the COVID-19-EVs-induced NET formation, we analyzed the expression profiles of microRNA (miRNAs) in COVID-19-EVs. We identified eight highly expressed miRNAs in COVID-19-EVs and explored their potential roles in COVID-19-EVs-mediated NET formation. Additionally, we explored the role of miR-20b-5p in COVID-19-EVs-induced NET formation.</p><p><strong>Results: </strong>In this study, we demonstrate that patients with COVID-19 have a higher concentration of serum EVs (COVID-19-EVs) than healthy controls (Normal-EVs). We also found that COVID-19-EVs are internalized by neutrophils to induced NET formation. Through comprehensive miRNA profiling of COVID-19-EVs versus Normal-EVs, we identified 78 differentially expressed miRNAs, with 27 of these being upregulated and 51 being downregulated. Subsequently, we discovered that COVID-19-EVs that were highly abundant with certain miRNAs promote NET formation. Specifically, miR-20b-5p was found to be the strongest inducer of NET formation of the identified miRNAs. Inhibition of miR-20b-5p resulted in a significant decrease in COVID-19-EVs-mediated induction of NET formation.</p><p><strong>Conclusion: </strong>Herein, we reveal a previously unknown role of COVID-19-EVs in NET formation, which contributes to COVID-19 progression. This study suggests that miR-20b-5p may serve as a potential therapeutic target for COVID-19 treatment.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"93"},"PeriodicalIF":8.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143442989","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cooperative Hedgehog/GLI and JAK/STAT signaling drives immunosuppressive tryptophan/kynurenine metabolism via synergistic induction of IDO1 in skin cancer.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1186/s12964-025-02101-6
Dominik P Elmer, Georg Stockmaier, Sandra Grund-Gröschke, Victoria Strobl, Hieu-Hoa Dang, Markus Wiederstein, David Licha, Anna Strobl, Anna Eglseer, Christina Sternberg, Suzana Tesanovic, Wolfgang Gruber, Florian Wolff, Richard Moriggl, Angela Risch, Roland Reischl, Christian G Huber, Peter W Krenn, Nikolaus Fortelny, Jutta Horejs-Hoeck, Fritz Aberger

Background: Pharmacological targeting of Hedgehog (HH)/GLI has proven effective for certain blood, brain and skin cancers including basal cell carcinoma (BCC). However, limited response rates and the development of drug resistance call for improved anti-HH therapies that take synergistic crosstalk mechanisms and immune evasion strategies into account. In previous work, we demonstrated that cooperation of HH/GLI and Interleukin 6 (IL6)/STAT3 signaling drives BCC growth. Whether synergistic HH-IL6 signaling promotes BCC via the activation of immune evasion mechanisms remained unclear.

Methods: HH-IL6 regulated immunosuppressive genes such as indoleamine 2,3-dioxygenase 1 (IDO1) were identified by gene expression profiling. IDO1 expression was evaluated in human BCC and melanoma models by qPCR and Western blot analyses. The cis-regulatory region of IDO1 was interrogated for HH-IL6-regulated GLI and STAT transcription factor binding and epigenetic modifications by targeted chromatin-immunoprecipitation and bisulfite pyrosequencing. Functional analyses of the immunosuppressive effects of IDO1 involved HPLC-MS measurements of its metabolites and the assessment of T cell proliferation via flow cytometry. Bioinformatic analyses of GLI-STAT cooperation were conducted on published bulk and single-cell RNA-seq data of human BCC and melanoma patients.

Results: We identified IDO1 as a target gene of cooperative GLI-STAT activity in BCC and melanoma. GLI1 and STAT3 transcription factors synergistically enhanced IDO1 expression by jointly binding to the cis-regulatory region of IDO1 and by increasing active chromatin marks at the histone level. In human melanoma cells, inhibition of GLI1 expression prevented the induction of IDO1 expression in response to IL6/STAT3 and IFNγ/STAT1 signaling. Pharmacological targeting of HH/GLI signaling reduced IDO1 expression, resulting in decreased production of the immunosuppressive metabolite kynurenine. Further, inhibition of GLI1 enhanced the efficacy of the selective IDO1 inhibitor epacadostat and rescued T cell proliferation by attenuating IDO1/kynurenine-mediated immunosuppression. Elevated expression of IDO1 correlated with active HH/GLI and JAK/STAT signaling in skin cancer patients supporting the clinical relevance of the mechanistic data presented.

Conclusions: These results identify the immunosuppressive IDO1-kynurenine pathway as a novel pro-tumorigenic target of oncogenic GLI and STAT1/STAT3 cooperation. Our data suggest simultaneous pharmacological targeting of these signaling axes as rational combination therapy in melanoma and non-melanoma skin cancers.

{"title":"Cooperative Hedgehog/GLI and JAK/STAT signaling drives immunosuppressive tryptophan/kynurenine metabolism via synergistic induction of IDO1 in skin cancer.","authors":"Dominik P Elmer, Georg Stockmaier, Sandra Grund-Gröschke, Victoria Strobl, Hieu-Hoa Dang, Markus Wiederstein, David Licha, Anna Strobl, Anna Eglseer, Christina Sternberg, Suzana Tesanovic, Wolfgang Gruber, Florian Wolff, Richard Moriggl, Angela Risch, Roland Reischl, Christian G Huber, Peter W Krenn, Nikolaus Fortelny, Jutta Horejs-Hoeck, Fritz Aberger","doi":"10.1186/s12964-025-02101-6","DOIUrl":"https://doi.org/10.1186/s12964-025-02101-6","url":null,"abstract":"<p><strong>Background: </strong>Pharmacological targeting of Hedgehog (HH)/GLI has proven effective for certain blood, brain and skin cancers including basal cell carcinoma (BCC). However, limited response rates and the development of drug resistance call for improved anti-HH therapies that take synergistic crosstalk mechanisms and immune evasion strategies into account. In previous work, we demonstrated that cooperation of HH/GLI and Interleukin 6 (IL6)/STAT3 signaling drives BCC growth. Whether synergistic HH-IL6 signaling promotes BCC via the activation of immune evasion mechanisms remained unclear.</p><p><strong>Methods: </strong>HH-IL6 regulated immunosuppressive genes such as indoleamine 2,3-dioxygenase 1 (IDO1) were identified by gene expression profiling. IDO1 expression was evaluated in human BCC and melanoma models by qPCR and Western blot analyses. The cis-regulatory region of IDO1 was interrogated for HH-IL6-regulated GLI and STAT transcription factor binding and epigenetic modifications by targeted chromatin-immunoprecipitation and bisulfite pyrosequencing. Functional analyses of the immunosuppressive effects of IDO1 involved HPLC-MS measurements of its metabolites and the assessment of T cell proliferation via flow cytometry. Bioinformatic analyses of GLI-STAT cooperation were conducted on published bulk and single-cell RNA-seq data of human BCC and melanoma patients.</p><p><strong>Results: </strong>We identified IDO1 as a target gene of cooperative GLI-STAT activity in BCC and melanoma. GLI1 and STAT3 transcription factors synergistically enhanced IDO1 expression by jointly binding to the cis-regulatory region of IDO1 and by increasing active chromatin marks at the histone level. In human melanoma cells, inhibition of GLI1 expression prevented the induction of IDO1 expression in response to IL6/STAT3 and IFNγ/STAT1 signaling. Pharmacological targeting of HH/GLI signaling reduced IDO1 expression, resulting in decreased production of the immunosuppressive metabolite kynurenine. Further, inhibition of GLI1 enhanced the efficacy of the selective IDO1 inhibitor epacadostat and rescued T cell proliferation by attenuating IDO1/kynurenine-mediated immunosuppression. Elevated expression of IDO1 correlated with active HH/GLI and JAK/STAT signaling in skin cancer patients supporting the clinical relevance of the mechanistic data presented.</p><p><strong>Conclusions: </strong>These results identify the immunosuppressive IDO1-kynurenine pathway as a novel pro-tumorigenic target of oncogenic GLI and STAT1/STAT3 cooperation. Our data suggest simultaneous pharmacological targeting of these signaling axes as rational combination therapy in melanoma and non-melanoma skin cancers.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"91"},"PeriodicalIF":8.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143442986","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Adenosine A2A receptor-mediated interactions between Th1+ T cells and the choroid plexus epithelium via IFN-γ signalling control T-Cell infiltration in experimental autoimmune encephalomyelitis.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1186/s12964-025-02100-7
Chenxing Qi, Yuwen Yang, Ping Tang, Cheng Zheng, Xuhang Li, Nan Jiang, Jia Qu, Jiang-Fan Chen, Wu Zheng

Background: Adenosine A2A receptor (A2AR) antagonists have been consistently demonstrated to protect against multiple sclerosis (MS) pathology, but A2AR knockout (A2AR-/-) mice exhibit exacerbated immune injury, raising concerns regarding the use of A2AR antagonists for MS treatment. Here, we revealed the critical involvement of A2AR-mediated interactions between Th1+ T cells and the choroid plexus (ChP) epithelium in the pathology of experimental autoimmune encephalomyelitis (EAE).

Methods: We assessed the effects of A2AR knockout on ChP gateway activity and the interferon gamma (IFN-γ)-secreting capacity of Th1+ T cells in an EAE model by immunofluorescence, qPCR and flow cytometry (FCM). We also investigated the effects of A2AR-mediated interactions between Th1+ T cells and the ChP epithelium on ChP gateway activity in vivo via intracerebroventricular (ICV) injection of Th1+ T cells and in vitro via coculture of ChP epithelial cells and splenic Th1+ T cells. We further knocked down IFN-γ receptor 1 (IFNGR1) specifically in the ChP of A2AR-/- mice via ICV injection of AAV2/5-shRNA (IFNGR1) to disrupt the interactions between Th1+ T cells and the ChP epithelium and thus assess the roles of these interactions in the development of EAE pathology.

Results: A2AR knockout disrupted the ChP barrier and increased T-cell infiltration across the ChP in EAE model mice. Coculture of splenic Th1+ T cells and ChP epithelial cells revealed that A2AR knockout in ChP epithelial cells strengthened the ChP barrier and attenuated T-cell migration, whereas A2AR knockout in Th1+ T cells increased the accumulation of Th1+ T cells in the ChP via the secretion of IFN-γ. Consistent with the coculture results, ICV injection of activated splenic Th1+ T cells from A2AR-/- mice increased the accumulation of T cells in the ChP to a greater extent than did injection of Th1+ T cells from A2AR+/+ mice. This effect was due to the increased secretion of IFN-γ in A2AR-/- mice compared with A2AR+/+ mice. Finally, ChP-specific knockdown of IFNGR1 attenuated A2AR knockout-induced T-cell infiltration, brain inflammation and EAE pathology.

Conclusion: A2AR-mediated interactions between Th1+ T cells and the ChP epithelium via the secretion of IFN-γ from CD4+ T cells and the binding IFN-γ to IFNGR1 in the ChP epithelium control immune cell invasion and the development of EAE pathology in A2AR-/- mice.

{"title":"Adenosine A<sub>2A</sub> receptor-mediated interactions between Th1<sup>+</sup> T cells and the choroid plexus epithelium via IFN-γ signalling control T-Cell infiltration in experimental autoimmune encephalomyelitis.","authors":"Chenxing Qi, Yuwen Yang, Ping Tang, Cheng Zheng, Xuhang Li, Nan Jiang, Jia Qu, Jiang-Fan Chen, Wu Zheng","doi":"10.1186/s12964-025-02100-7","DOIUrl":"https://doi.org/10.1186/s12964-025-02100-7","url":null,"abstract":"<p><strong>Background: </strong>Adenosine A<sub>2A</sub> receptor (A<sub>2A</sub>R) antagonists have been consistently demonstrated to protect against multiple sclerosis (MS) pathology, but A<sub>2A</sub>R knockout (A<sub>2A</sub>R<sup>-/-</sup>) mice exhibit exacerbated immune injury, raising concerns regarding the use of A<sub>2A</sub>R antagonists for MS treatment. Here, we revealed the critical involvement of A<sub>2A</sub>R-mediated interactions between Th1<sup>+</sup> T cells and the choroid plexus (ChP) epithelium in the pathology of experimental autoimmune encephalomyelitis (EAE).</p><p><strong>Methods: </strong>We assessed the effects of A<sub>2A</sub>R knockout on ChP gateway activity and the interferon gamma (IFN-γ)-secreting capacity of Th1<sup>+</sup> T cells in an EAE model by immunofluorescence, qPCR and flow cytometry (FCM). We also investigated the effects of A<sub>2A</sub>R-mediated interactions between Th1<sup>+</sup> T cells and the ChP epithelium on ChP gateway activity in vivo via intracerebroventricular (ICV) injection of Th1<sup>+</sup> T cells and in vitro via coculture of ChP epithelial cells and splenic Th1<sup>+</sup> T cells. We further knocked down IFN-γ receptor 1 (IFNGR1) specifically in the ChP of A<sub>2A</sub>R<sup>-/-</sup> mice via ICV injection of AAV2/5-shRNA (IFNGR1) to disrupt the interactions between Th1<sup>+</sup> T cells and the ChP epithelium and thus assess the roles of these interactions in the development of EAE pathology.</p><p><strong>Results: </strong>A<sub>2A</sub>R knockout disrupted the ChP barrier and increased T-cell infiltration across the ChP in EAE model mice. Coculture of splenic Th1<sup>+</sup> T cells and ChP epithelial cells revealed that A<sub>2A</sub>R knockout in ChP epithelial cells strengthened the ChP barrier and attenuated T-cell migration, whereas A<sub>2A</sub>R knockout in Th1<sup>+</sup> T cells increased the accumulation of Th1<sup>+</sup> T cells in the ChP via the secretion of IFN-γ. Consistent with the coculture results, ICV injection of activated splenic Th1<sup>+</sup> T cells from A<sub>2A</sub>R<sup>-/-</sup> mice increased the accumulation of T cells in the ChP to a greater extent than did injection of Th1<sup>+</sup> T cells from A<sub>2A</sub>R<sup>+/+</sup> mice. This effect was due to the increased secretion of IFN-γ in A<sub>2A</sub>R<sup>-/-</sup> mice compared with A<sub>2A</sub>R<sup>+/+</sup> mice. Finally, ChP-specific knockdown of IFNGR1 attenuated A<sub>2A</sub>R knockout-induced T-cell infiltration, brain inflammation and EAE pathology.</p><p><strong>Conclusion: </strong>A<sub>2A</sub>R-mediated interactions between Th1<sup>+</sup> T cells and the ChP epithelium via the secretion of IFN-γ from CD4<sup>+</sup> T cells and the binding IFN-γ to IFNGR1 in the ChP epithelium control immune cell invasion and the development of EAE pathology in A<sub>2A</sub>R<sup>-/-</sup> mice.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"94"},"PeriodicalIF":8.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143442984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IgA displays site- and subclass-specific glycoform differences despite equal glycoenzyme expression.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1186/s12964-025-02088-0
David Falck, Maria V Sokolova, Carolien A M Koeleman, Vanessa Irumva, Philipp Kirchner, Sebastian R Schulz, Katja G Schmidt, Thomas Harrer, Arif B Ekici, Bernd Spriewald, Georg Schett, Manfred Wuhrer, Martin Herrmann, Ulrike Steffen

Background: Glycosylation is an important posttranslational modification of proteins and in most cases indispensable for proper protein function. Like most soluble proteins, IgA, the second most prevalent antibody in human serum, contains several N- and O-glycosylation sites. While for IgG the impact of Fc glycosylation on effector functions and inflammatory potential has been studied intensively, only little is known for IgA. In addition, only glimpses exist regarding the regulation of IgA glycosylation. We have previously shown that IgA1 and IgA2 differ functionally and also show differences in their glycosylation pattern. The more pro-inflammatory IgA2 which is linked to autoimmune diseases displays decreased sialylation, galactosylation, fucosylation and bisection as compared to IgA1. In the present study, we aimed to investigate these differences in glycosylation in detail and to explore the mechanisms underlying them.

Methods: IgA1 and IgA2 was isolated from serum of 12 healthy donors. Site specific glycosylation was analyzed by mass spectrometry. In addition, human bone marrow plasma cells were investigated using single cell mRNA sequencing, flow cytometry and ELISpot.

Results: We found that certain glycoforms greatly differ in their abundance between IgA1 and IgA2 while others are equally abundant. Overall, the IgA2 glycans displayed a more immature phenotype with a higher prevalence of oligomannose and fewer fully processed glycans. Of note, these differences can't be explained by differences in the glycosylation enzyme machinery as mRNA sequencing and flow cytometry analysis showed equal enzyme expression in IgA1 and IgA2 producing plasma cells. ELISpot analysis suggested a slightly increased antibody production rate in IgA2 producing plasma cells which might contribute to its lower glycan processing rates. But this difference was only minor, suggesting that further factors such as steric accessibility determine glycan processing. This is supported by the fact that glycans at different positions on the same IgA chain differ dramatically in fucosylation, sialylation and bisection.

Conclusion: In summary, our detailed overview of IgA1 and IgA2 glycosylation shows a class, subclass, and site-specific glycosylation fingerprint, most likely due to structural differences of the protein backbones.

{"title":"IgA displays site- and subclass-specific glycoform differences despite equal glycoenzyme expression.","authors":"David Falck, Maria V Sokolova, Carolien A M Koeleman, Vanessa Irumva, Philipp Kirchner, Sebastian R Schulz, Katja G Schmidt, Thomas Harrer, Arif B Ekici, Bernd Spriewald, Georg Schett, Manfred Wuhrer, Martin Herrmann, Ulrike Steffen","doi":"10.1186/s12964-025-02088-0","DOIUrl":"https://doi.org/10.1186/s12964-025-02088-0","url":null,"abstract":"<p><strong>Background: </strong>Glycosylation is an important posttranslational modification of proteins and in most cases indispensable for proper protein function. Like most soluble proteins, IgA, the second most prevalent antibody in human serum, contains several N- and O-glycosylation sites. While for IgG the impact of Fc glycosylation on effector functions and inflammatory potential has been studied intensively, only little is known for IgA. In addition, only glimpses exist regarding the regulation of IgA glycosylation. We have previously shown that IgA1 and IgA2 differ functionally and also show differences in their glycosylation pattern. The more pro-inflammatory IgA2 which is linked to autoimmune diseases displays decreased sialylation, galactosylation, fucosylation and bisection as compared to IgA1. In the present study, we aimed to investigate these differences in glycosylation in detail and to explore the mechanisms underlying them.</p><p><strong>Methods: </strong>IgA1 and IgA2 was isolated from serum of 12 healthy donors. Site specific glycosylation was analyzed by mass spectrometry. In addition, human bone marrow plasma cells were investigated using single cell mRNA sequencing, flow cytometry and ELISpot.</p><p><strong>Results: </strong>We found that certain glycoforms greatly differ in their abundance between IgA1 and IgA2 while others are equally abundant. Overall, the IgA2 glycans displayed a more immature phenotype with a higher prevalence of oligomannose and fewer fully processed glycans. Of note, these differences can't be explained by differences in the glycosylation enzyme machinery as mRNA sequencing and flow cytometry analysis showed equal enzyme expression in IgA1 and IgA2 producing plasma cells. ELISpot analysis suggested a slightly increased antibody production rate in IgA2 producing plasma cells which might contribute to its lower glycan processing rates. But this difference was only minor, suggesting that further factors such as steric accessibility determine glycan processing. This is supported by the fact that glycans at different positions on the same IgA chain differ dramatically in fucosylation, sialylation and bisection.</p><p><strong>Conclusion: </strong>In summary, our detailed overview of IgA1 and IgA2 glycosylation shows a class, subclass, and site-specific glycosylation fingerprint, most likely due to structural differences of the protein backbones.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"92"},"PeriodicalIF":8.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143442991","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Microglial depletion decreases Müller cell maturation and inner retinal vascular density.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-17 DOI: 10.1186/s12964-025-02083-5
Nathaniel Rowthorn-Apel, Naveen Vridhachalam, Kip M Connor, Gracia M Bonilla, Ruslan Sadreyev, Charandeep Singh, Gopalan Gnanaguru

Background: The neuroretinal vascular system is comprised of three interconnected layers. The initial superficial vascular plexus formation is guided by astrocytes around birth in mice. The formation of the deep and intermediate vascular plexuses occurs in the second postnatal week and is driven by Müller-cell-derived angiogenic signaling. Previously, we reported that microglia play an important role in regulating astrocyte density during superficial vascular plexus formation. Here, we investigated the role of microglia in regulating Müller-cell-dependent inner retinal vascular development.

Methodology: In this study, we depleted microglia during retinal development using Csf1R antagonist (PLX5622). We characterized the developmental progression of inner retinal vascular growth, effect of microglial depletion on inner retinal vascular growth and Müller cell marker expressions by immunostaining. Differential expressions of genes in the control and microglia depleted groups were analyzed by mRNA-seq and qPCR. Unpaired t-test was performed to determine the statistical differences between groups.

Results: This study show that microglia interact with Müller cells and the growing inner retinal vasculature. Depletion of microglia resulted in reduced inner retinal vascular layers densities and decreased Vegfa isoforms transcript levels. RNA-seq analysis further revealed that microglial depletion significantly reduced specific Müller cell maturation markers including glutamine synthetase, responsible for glutamine biosynthesis, necessary for angiogenesis.

Conclusions: Our study reveals an important role for microglia in facilitating inner retinal angiogenesis and Müller cell maturation.

{"title":"Microglial depletion decreases Müller cell maturation and inner retinal vascular density.","authors":"Nathaniel Rowthorn-Apel, Naveen Vridhachalam, Kip M Connor, Gracia M Bonilla, Ruslan Sadreyev, Charandeep Singh, Gopalan Gnanaguru","doi":"10.1186/s12964-025-02083-5","DOIUrl":"https://doi.org/10.1186/s12964-025-02083-5","url":null,"abstract":"<p><strong>Background: </strong>The neuroretinal vascular system is comprised of three interconnected layers. The initial superficial vascular plexus formation is guided by astrocytes around birth in mice. The formation of the deep and intermediate vascular plexuses occurs in the second postnatal week and is driven by Müller-cell-derived angiogenic signaling. Previously, we reported that microglia play an important role in regulating astrocyte density during superficial vascular plexus formation. Here, we investigated the role of microglia in regulating Müller-cell-dependent inner retinal vascular development.</p><p><strong>Methodology: </strong>In this study, we depleted microglia during retinal development using Csf1R antagonist (PLX5622). We characterized the developmental progression of inner retinal vascular growth, effect of microglial depletion on inner retinal vascular growth and Müller cell marker expressions by immunostaining. Differential expressions of genes in the control and microglia depleted groups were analyzed by mRNA-seq and qPCR. Unpaired t-test was performed to determine the statistical differences between groups.</p><p><strong>Results: </strong>This study show that microglia interact with Müller cells and the growing inner retinal vasculature. Depletion of microglia resulted in reduced inner retinal vascular layers densities and decreased Vegfa isoforms transcript levels. RNA-seq analysis further revealed that microglial depletion significantly reduced specific Müller cell maturation markers including glutamine synthetase, responsible for glutamine biosynthesis, necessary for angiogenesis.</p><p><strong>Conclusions: </strong>Our study reveals an important role for microglia in facilitating inner retinal angiogenesis and Müller cell maturation.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"90"},"PeriodicalIF":8.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143442993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting membrane contact sites to mediate lipid dynamics: innovative cancer therapies.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-15 DOI: 10.1186/s12964-025-02089-z
Jie Wang, Meifeng Wang, Xueni Zeng, Yanhan Li, Lingzhi Lei, Changan Chen, Xi Lin, Peiyuan Fang, Yuxuan Guo, Xianjie Jiang, Yian Wang, Lihong Chen, Jun Long

Membrane contact sites (MCS) are specialized regions where organelles are closely interconnected through membrane structures, facilitating the transfer and exchange of ions, lipids, and other molecules. This proximity enables a synergistic regulation of cellular homeostasis and functions. The formation and maintenance of these contact sites are governed by specific proteins that bring organelle membranes into close apposition, thereby enabling functional crosstalk between cellular compartments. In eukaryotic cells, lipids are primarily synthesized and metabolized within distinct organelles and must be transported through MCS to ensure proper cellular function. Consequently, MCS act as pivotal platforms for lipid synthesis and trafficking, particularly in cancer cells and immune cells within the tumor microenvironment, where dynamic alterations are critical for maintaining lipid homeostasis. This article provides a comprehensive analysis of how these cells exploit membrane contact sites to modulate lipid synthesis, metabolism, and transport, with a specific focus on how MCS-mediated lipid dynamics influence tumor progression. We also examine the differences in MCS and associated molecules across various cancer types, exploring novel therapeutic strategies targeting MCS-related lipid metabolism for the development of anticancer drugs, while also addressing the challenges involved.

{"title":"Targeting membrane contact sites to mediate lipid dynamics: innovative cancer therapies.","authors":"Jie Wang, Meifeng Wang, Xueni Zeng, Yanhan Li, Lingzhi Lei, Changan Chen, Xi Lin, Peiyuan Fang, Yuxuan Guo, Xianjie Jiang, Yian Wang, Lihong Chen, Jun Long","doi":"10.1186/s12964-025-02089-z","DOIUrl":"10.1186/s12964-025-02089-z","url":null,"abstract":"<p><p>Membrane contact sites (MCS) are specialized regions where organelles are closely interconnected through membrane structures, facilitating the transfer and exchange of ions, lipids, and other molecules. This proximity enables a synergistic regulation of cellular homeostasis and functions. The formation and maintenance of these contact sites are governed by specific proteins that bring organelle membranes into close apposition, thereby enabling functional crosstalk between cellular compartments. In eukaryotic cells, lipids are primarily synthesized and metabolized within distinct organelles and must be transported through MCS to ensure proper cellular function. Consequently, MCS act as pivotal platforms for lipid synthesis and trafficking, particularly in cancer cells and immune cells within the tumor microenvironment, where dynamic alterations are critical for maintaining lipid homeostasis. This article provides a comprehensive analysis of how these cells exploit membrane contact sites to modulate lipid synthesis, metabolism, and transport, with a specific focus on how MCS-mediated lipid dynamics influence tumor progression. We also examine the differences in MCS and associated molecules across various cancer types, exploring novel therapeutic strategies targeting MCS-related lipid metabolism for the development of anticancer drugs, while also addressing the challenges involved.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"89"},"PeriodicalIF":8.2,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830217/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143426665","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
A novel 5'tRNA-derived fragment tRF-Tyr inhibits tumor progression by targeting hnRNPD in gastric cancer.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-14 DOI: 10.1186/s12964-025-02086-2
Huaiping Cui, Zhaodong Liu, Lipan Peng, Lijun Liu, Xiaozhou Xie, Yudi Zhang, Zi Gao, Chi Zhang, Xinshuai Yu, Yonghao Hu, Jin Liu, Liang Shang, Leping Li

Background: Transfer RNA-derived small RNAs (tsRNAs), including tRNA-derived fragments (tRFs) and tRNA halves (tiRNAs), constitute a novel class of small noncoding RNAs (sncRNAs). tsRNAs have been linked to tumorigenesis and the progression of carcinogenesis; however, the precise molecular mechanism through which tRFs act in gastric cancer (GC) remains unknown.

Methods: tRF-Tyr is a potential GC tumor suppressor that was identified through high-throughput sequencing technology. The expression and subcellular localization of tRF-Tyr in GC were detected by via qRT‒PCR and FISH. RNA pull-down, mass spectrometry, RNA immunoprecipitation (RIP), dual-luciferase reporter and rescue assays were performed to explore the regulatory mechanisms through which tRF-Tyr acts in GC.

Results: tRF-Tyr was significantly downregulated and the downregulation of its mainly concentrated in the nuclei of GC cells. Functionally, tRF-Tyr inhibited the proliferation, invasiveness and migration of GC cells and promoted GC cells apoptosis in vitro; meanwhile, tRF-Tyr inhibited tumor growth in vivo. Mechanistically, tRF-Tyr bound directly to the hnRNPD protein and competitively inhibited the binding of hnRNPD to the c-Myc 3'UTR, thereby, regulating the c-Myc/Bcl2/Bax pathway and ultimately inhibiting the progression of GC.

Conclusions: This study focused on a novel GC suppressor, tRF-Tyr, and revealed a previously undiscovered mechanism that tRF-Tyr inhibits tumor progression by targeting hnRNPD. These findings provide new insight into the involvement of tRFs in GC and suggest a novel target for GC treatment.

{"title":"A novel 5'tRNA-derived fragment tRF-Tyr inhibits tumor progression by targeting hnRNPD in gastric cancer.","authors":"Huaiping Cui, Zhaodong Liu, Lipan Peng, Lijun Liu, Xiaozhou Xie, Yudi Zhang, Zi Gao, Chi Zhang, Xinshuai Yu, Yonghao Hu, Jin Liu, Liang Shang, Leping Li","doi":"10.1186/s12964-025-02086-2","DOIUrl":"10.1186/s12964-025-02086-2","url":null,"abstract":"<p><strong>Background: </strong>Transfer RNA-derived small RNAs (tsRNAs), including tRNA-derived fragments (tRFs) and tRNA halves (tiRNAs), constitute a novel class of small noncoding RNAs (sncRNAs). tsRNAs have been linked to tumorigenesis and the progression of carcinogenesis; however, the precise molecular mechanism through which tRFs act in gastric cancer (GC) remains unknown.</p><p><strong>Methods: </strong>tRF-Tyr is a potential GC tumor suppressor that was identified through high-throughput sequencing technology. The expression and subcellular localization of tRF-Tyr in GC were detected by via qRT‒PCR and FISH. RNA pull-down, mass spectrometry, RNA immunoprecipitation (RIP), dual-luciferase reporter and rescue assays were performed to explore the regulatory mechanisms through which tRF-Tyr acts in GC.</p><p><strong>Results: </strong>tRF-Tyr was significantly downregulated and the downregulation of its mainly concentrated in the nuclei of GC cells. Functionally, tRF-Tyr inhibited the proliferation, invasiveness and migration of GC cells and promoted GC cells apoptosis in vitro; meanwhile, tRF-Tyr inhibited tumor growth in vivo. Mechanistically, tRF-Tyr bound directly to the hnRNPD protein and competitively inhibited the binding of hnRNPD to the c-Myc 3'UTR, thereby, regulating the c-Myc/Bcl2/Bax pathway and ultimately inhibiting the progression of GC.</p><p><strong>Conclusions: </strong>This study focused on a novel GC suppressor, tRF-Tyr, and revealed a previously undiscovered mechanism that tRF-Tyr inhibits tumor progression by targeting hnRNPD. These findings provide new insight into the involvement of tRFs in GC and suggest a novel target for GC treatment.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"88"},"PeriodicalIF":8.2,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11829405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143426656","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
Dysregulation of neutrophil in sepsis: recent insights and advances. 脓毒症中的中性粒细胞失调:最新见解和进展。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-14 DOI: 10.1186/s12964-025-02098-y
Ji Zhang, Yuwen Shao, Jingyi Wu, Jing Zhang, Xiangsheng Xiong, Jingjing Mao, Yunwei Wei, Changhong Miao, Hao Zhang

Sepsis remains the leading cause of death in intensive care units. Despite newer antimicrobial and supportive therapies, specific treatments are still lacking. Neutrophils are pivotal components of the effector phase of the host immune defense against pathogens and play a crucial role in the control of infections under normal circumstances. In addition to its anti-infective effects, the dysregulation and overactivation of neutrophils may lead to severe inflammation or tissue damage and are potential mechanisms for poor prognosis in sepsis. This review focuses on recent advancements in the understanding of the functional status of neutrophils across various pathological stages of sepsis to explore the mechanisms by which neutrophils participate in sepsis progression and provide insights for the treatment of sepsis by targeting neutrophils.

{"title":"Dysregulation of neutrophil in sepsis: recent insights and advances.","authors":"Ji Zhang, Yuwen Shao, Jingyi Wu, Jing Zhang, Xiangsheng Xiong, Jingjing Mao, Yunwei Wei, Changhong Miao, Hao Zhang","doi":"10.1186/s12964-025-02098-y","DOIUrl":"10.1186/s12964-025-02098-y","url":null,"abstract":"<p><p>Sepsis remains the leading cause of death in intensive care units. Despite newer antimicrobial and supportive therapies, specific treatments are still lacking. Neutrophils are pivotal components of the effector phase of the host immune defense against pathogens and play a crucial role in the control of infections under normal circumstances. In addition to its anti-infective effects, the dysregulation and overactivation of neutrophils may lead to severe inflammation or tissue damage and are potential mechanisms for poor prognosis in sepsis. This review focuses on recent advancements in the understanding of the functional status of neutrophils across various pathological stages of sepsis to explore the mechanisms by which neutrophils participate in sepsis progression and provide insights for the treatment of sepsis by targeting neutrophils.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"87"},"PeriodicalIF":8.2,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11827254/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143426659","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
TNFAIP2 promotes NF-κB signaling mediate lymph node metastasis of oral squamous cell carcinoma by protecting IKKβ from ubiquitin proteasome degradation.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-13 DOI: 10.1186/s12964-025-02077-3
Teng Xu, Yaning Wang, Zechen Zhao, Jinsong Wang, Zhenyuan Zhao, Yuemei Yang, Xiaomeng Song, Qingguo Lai

Background: Tumor dissemination is a life-threatening event which confers to most cancer-related deaths with limited effective therapeutic option. TNFα-induced protein 2 (TNFAIP2) reveals pro-metastasis potential in several cancers. However, its definite role and underlying mechanism in oral squamous cell carcinoma (OSCC) is largely unknown.

Methods: The impact of TNFAIP2 on tumor metastasis was assessed based on the conditional knockout mouse with 4-nitroquinoline-1-oxide (4NQO) induced OSCC model through feature and immunohistochemistry analysis. To explore the specific mechanism, enrichment analysis and co-immunoprecipitation were applied. Meanwhile, the nano-hydroxyapatite (nHAp) and poly-L-lysine (PLL) based RNA interference delivery system was designed to restrict tumor dissemination.

Results: The conditional knockout Tnfaip2 in epithelium reduced tumor initiation rate, differentiation degree and cervical lymph node metastasis (LNM) in mouse exposed to 4NQO. Enrichment analysis suggested nuclear factor-kappa B (NF-κB) signaling was associated with these effects. Western blot proved that TNFAIP2 prevented the ubiquitin proteasome degradation of inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKKβ), a classical transcriptional activator protein in NF-κB signaling. Mechanistically, TNFAIP2 was demonstrated to competitively interact with kelch-like ECH-associated protein 1 (KEAP1) to avoid IKKβ from ubiquitination at K63 and proteasomal degradation subsequently, which finally sustained NF-κB signaling and facilitated tumor metastasis by enhancing epithelial-mesenchymal transition (EMT) and lymphangiogenesis. Notably, the synthetic small interfering RNA delivery systems nHAp@PLL-siTnfaip2 showed significant effect in attenuating tumor progression of OSCC mouse.

Conclusion: Above results showed TNFAIP2 promoted EMT and lymphangiogenesis of OSCC by regulating NF-κB signaling, a mechanism that was dependent on the interaction with KEAP1 competitively. The nHAp based TNFAIP2 interference might serve as a novel therapeutic in limiting OSCC metastasis.

{"title":"TNFAIP2 promotes NF-κB signaling mediate lymph node metastasis of oral squamous cell carcinoma by protecting IKKβ from ubiquitin proteasome degradation.","authors":"Teng Xu, Yaning Wang, Zechen Zhao, Jinsong Wang, Zhenyuan Zhao, Yuemei Yang, Xiaomeng Song, Qingguo Lai","doi":"10.1186/s12964-025-02077-3","DOIUrl":"10.1186/s12964-025-02077-3","url":null,"abstract":"<p><strong>Background: </strong>Tumor dissemination is a life-threatening event which confers to most cancer-related deaths with limited effective therapeutic option. TNFα-induced protein 2 (TNFAIP2) reveals pro-metastasis potential in several cancers. However, its definite role and underlying mechanism in oral squamous cell carcinoma (OSCC) is largely unknown.</p><p><strong>Methods: </strong>The impact of TNFAIP2 on tumor metastasis was assessed based on the conditional knockout mouse with 4-nitroquinoline-1-oxide (4NQO) induced OSCC model through feature and immunohistochemistry analysis. To explore the specific mechanism, enrichment analysis and co-immunoprecipitation were applied. Meanwhile, the nano-hydroxyapatite (nHAp) and poly-L-lysine (PLL) based RNA interference delivery system was designed to restrict tumor dissemination.</p><p><strong>Results: </strong>The conditional knockout Tnfaip2 in epithelium reduced tumor initiation rate, differentiation degree and cervical lymph node metastasis (LNM) in mouse exposed to 4NQO. Enrichment analysis suggested nuclear factor-kappa B (NF-κB) signaling was associated with these effects. Western blot proved that TNFAIP2 prevented the ubiquitin proteasome degradation of inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKKβ), a classical transcriptional activator protein in NF-κB signaling. Mechanistically, TNFAIP2 was demonstrated to competitively interact with kelch-like ECH-associated protein 1 (KEAP1) to avoid IKKβ from ubiquitination at K63 and proteasomal degradation subsequently, which finally sustained NF-κB signaling and facilitated tumor metastasis by enhancing epithelial-mesenchymal transition (EMT) and lymphangiogenesis. Notably, the synthetic small interfering RNA delivery systems nHAp@PLL-siTnfaip2 showed significant effect in attenuating tumor progression of OSCC mouse.</p><p><strong>Conclusion: </strong>Above results showed TNFAIP2 promoted EMT and lymphangiogenesis of OSCC by regulating NF-κB signaling, a mechanism that was dependent on the interaction with KEAP1 competitively. The nHAp based TNFAIP2 interference might serve as a novel therapeutic in limiting OSCC metastasis.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"83"},"PeriodicalIF":8.2,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11827437/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143416311","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
Biogenesis of stress granules and their role in the regulation of stress-induced male reproduction disorders.
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-02-13 DOI: 10.1186/s12964-025-02054-w
Jiaxin Li, Linyuan Shen, Kai Wang, Shuang Wu, Yan Wang, Yuheng Pan, Siyu Chen, Ting Zhao, Ye Zhao, Lili Niu, Lei Chen, Shunhua Zhang, Li Zhu, Mailin Gan

Stress granules (SGs) are conserved messenger ribonucleoprotein (mRNP) granules that form through rapid coalescence in the cytoplasm of eukaryotic cells under stressful environments. These dynamic membrane-free organelles can respond to a variety of both intracellular and extracellular stressors. Studies have shown that stress conditions such as heat stress, arsenite exposure, and hypoxic stress can induce SGs formation. The formation of SGs helps mitigates the effects of environmental stimuli on cells, protects them from damage, and promotes cell survival. This paper focuses on the biogenesis of SGs and summarizes the role in regulating environmental stress-induced male reproductive disorders, with the aim of exploring SGs as a potential means of mitigating male reproduction disorders. Numerous studies have demonstrated that the detrimental effects of environmental stress on germ cells can be effectively suppressed by regulating the formation and timely disassembly of SGs. Therefore, regulating the phosphorylation of eIF2α and the assembly and disassembly of SGs could offer a promising therapeutic strategy to alleviate the impacts of environmental stress on male reproduction health.

{"title":"Biogenesis of stress granules and their role in the regulation of stress-induced male reproduction disorders.","authors":"Jiaxin Li, Linyuan Shen, Kai Wang, Shuang Wu, Yan Wang, Yuheng Pan, Siyu Chen, Ting Zhao, Ye Zhao, Lili Niu, Lei Chen, Shunhua Zhang, Li Zhu, Mailin Gan","doi":"10.1186/s12964-025-02054-w","DOIUrl":"10.1186/s12964-025-02054-w","url":null,"abstract":"<p><p>Stress granules (SGs) are conserved messenger ribonucleoprotein (mRNP) granules that form through rapid coalescence in the cytoplasm of eukaryotic cells under stressful environments. These dynamic membrane-free organelles can respond to a variety of both intracellular and extracellular stressors. Studies have shown that stress conditions such as heat stress, arsenite exposure, and hypoxic stress can induce SGs formation. The formation of SGs helps mitigates the effects of environmental stimuli on cells, protects them from damage, and promotes cell survival. This paper focuses on the biogenesis of SGs and summarizes the role in regulating environmental stress-induced male reproductive disorders, with the aim of exploring SGs as a potential means of mitigating male reproduction disorders. Numerous studies have demonstrated that the detrimental effects of environmental stress on germ cells can be effectively suppressed by regulating the formation and timely disassembly of SGs. Therefore, regulating the phosphorylation of eIF2α and the assembly and disassembly of SGs could offer a promising therapeutic strategy to alleviate the impacts of environmental stress on male reproduction health.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"84"},"PeriodicalIF":8.2,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11827146/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143416306","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