Long non-coding RNAs(lncRNAs)play key roles in colorectal carcinogenesis.Here,we aimed to identify the risk SNP-induced lncRNAs and to investigate their roles in colorectal carcinogenesis.First,we identified rs6695584...Long non-coding RNAs(lncRNAs)play key roles in colorectal carcinogenesis.Here,we aimed to identify the risk SNP-induced lncRNAs and to investigate their roles in colorectal carcinogenesis.First,we identified rs6695584 as the causative SNP in 1 q41 locus.The A>G mutation of rs6695584 created a protein-binding motif of BATF,altered the enhancer activity,and subsequently activated IncSLCCl expression.Further validation in two independent CRC cohorts confirmed the upregulation of IncSLCCl in CRC tissues,and revealed that increased IncSLCCl expression was associated with poor survival in CRC patients.Mechanistically,lncRNA-SLCCl interacted with AHR and transcriptionally activated HK2 expression,the crucial enzyme in glucose metabolism,thereby driving the glycolysis pathway and accelerating CRC tumor growth.The functional assays revealed that IncSLCCl induced glycolysis activation and tumor growth in CRC mediated by HK2.In addition,HK2 was upregulated in colorectal cancer tissues and positively correlated with IncSLCCl expression and patient survival.Taken together,our findings reveal a risk SNP-mediated oncogene lncRNA-SLCCl promotes CRC through activating the glycolysis pathway.展开更多
As one of the best characterized adult stem cells,hematopoietic stem cell(HSC)homeostasis is of great importance to hematopoiesis and immunity due to HSC’s abilities of self-renewal and multi-lineage differentiation ...As one of the best characterized adult stem cells,hematopoietic stem cell(HSC)homeostasis is of great importance to hematopoiesis and immunity due to HSC’s abilities of self-renewal and multi-lineage differentiation into functional blood cells.However,excessive self-renewal of HSCs can lead to severe hematopoietic malignancies like leukemia,whereas deficient selfrenewal of HSCs may result in HSC exhaustion and eventually apoptosis of specialized cells,giving rise to abnormalities such as immunodeficiency or anemia.How HSC homeostasis is maintained has been studied for decades and regulatory factors can be generally categorized into two classes:genetic factors and epigenetic factors.Although genetic factors such as signaling pathways or transcription factors have been well explored,recent studies have emerged the indispensable roles of epigenetic factors.In this review,we have summarized regulatory mechanisms of HSC homeostasis by epigenetic factors,including DNA methylation,histone modification,chromatin remodeling,non-coding RNAs,and RNA modification,which will facilitate applications such as HSC ex vivo expansion and exploration of novel therapeutic approaches for many hematological diseases.展开更多
Dear Editor,COVID-19(Coronavirus Disease 2019)is causing an unprecedented public health crisis.Protein translation is crucial for virus lifecycle.Nucleocapsid(N)protein is among the most abundant SARS-CoV-2 proteins a...Dear Editor,COVID-19(Coronavirus Disease 2019)is causing an unprecedented public health crisis.Protein translation is crucial for virus lifecycle.Nucleocapsid(N)protein is among the most abundant SARS-CoV-2 proteins and highly conserved across coronavirus genus.1 However,its function in subverting host translation machinery is still elusive.展开更多
Clinical evidence supports the notion that T cell exhaustion and terminal differentiation pose challenges to the persistence and effectiveness of chimeric antigen receptor-T(CAR-T)cells.MEK1/2 inhibitors(MEKIs),widely...Clinical evidence supports the notion that T cell exhaustion and terminal differentiation pose challenges to the persistence and effectiveness of chimeric antigen receptor-T(CAR-T)cells.MEK1/2 inhibitors(MEKIs),widely used in cancer treatment due to their ability to inhibit aberrant MAPK signaling,have shown potential synergistic effects when combined with immunotherapy.However,the impact and mechanisms of MEKIs on CAR-T cells remain uncertain and controversial.To address this,we conducted a comprehensive investigation to determine whether MEKIs enhance or impair the efficacy of CAR-T cells.Our findings revealed that MEKIs attenuated CAR-T cell exhaustion and terminal differentiation induced by tonic signaling and antigen stimulation,thereby improving CAR-T cell efficacy against hematological and solid tumors.Remarkably,these effects were independent of the specific scFvs and costimulatory domains utilized in CARs.Mechanistically,analysis of bulk and single-cell transcriptional profiles demonstrates that the effect of MEK inhibition was related to diminish anabolic metabolism and downregulation of c-Fos and JunB.Additionally,the overexpression of c-Fos or JunB in CAR-T cells counteracted the effects of MEK inhibition.Furthermore,our Cut-and-Tag assay revealed that MEK inhibition downregulated the JunB-driven gene profiles associated with exhaustion,differentiation,anergy,glycolysis,and apoptosis.In summary,our research unveil the critical role of the MAPK-c-Fos-JunB axis in driving CAR-T cell exhaustion and terminal differentiation.These mechanistic insights significantly broaden the potential application of MEKIs to enhance the effectiveness of CAR-T therapy.展开更多
基金supported in part by grants from the State Key R&D Program(2020YFA0509200)the National Natural Science Foundation of China(81421001,81530072,81830081,81871901,81874159,81902368,31970718,81770165)+4 种基金Shanghai Municipal Health Commission,Collaborative Innovation Cluster Project(2019CXJQ02)"Shu Guang"project supported by Shanghai Municipal Education Commission and Shanghai Education Developm ent Foundation(17SG18)the Program for Professor of Special Appointm ent(Eastern Scholar No.201268 and 2015 Youth Eastern Scholar No.QD2015003)at Shanghai Institutions of Higher LearningShanghai Municipal Education Commission—Gaofeng Clinical Medicine Grant Support(No.20152512,20161309)Innovative research team of high-level local universities in Shanghai.
文摘Long non-coding RNAs(lncRNAs)play key roles in colorectal carcinogenesis.Here,we aimed to identify the risk SNP-induced lncRNAs and to investigate their roles in colorectal carcinogenesis.First,we identified rs6695584 as the causative SNP in 1 q41 locus.The A>G mutation of rs6695584 created a protein-binding motif of BATF,altered the enhancer activity,and subsequently activated IncSLCCl expression.Further validation in two independent CRC cohorts confirmed the upregulation of IncSLCCl in CRC tissues,and revealed that increased IncSLCCl expression was associated with poor survival in CRC patients.Mechanistically,lncRNA-SLCCl interacted with AHR and transcriptionally activated HK2 expression,the crucial enzyme in glucose metabolism,thereby driving the glycolysis pathway and accelerating CRC tumor growth.The functional assays revealed that IncSLCCl induced glycolysis activation and tumor growth in CRC mediated by HK2.In addition,HK2 was upregulated in colorectal cancer tissues and positively correlated with IncSLCCl expression and patient survival.Taken together,our findings reveal a risk SNP-mediated oncogene lncRNA-SLCCl promotes CRC through activating the glycolysis pathway.
基金supported by grants from the National Key R&D Program of China,Stem Cell and Translation Research(2018YFA0109300)Zhejiang Province Science Foundation for Distinguished Young Scholars(LR19H080001)the National Natural Science Foundation of China(81870080).
文摘As one of the best characterized adult stem cells,hematopoietic stem cell(HSC)homeostasis is of great importance to hematopoiesis and immunity due to HSC’s abilities of self-renewal and multi-lineage differentiation into functional blood cells.However,excessive self-renewal of HSCs can lead to severe hematopoietic malignancies like leukemia,whereas deficient selfrenewal of HSCs may result in HSC exhaustion and eventually apoptosis of specialized cells,giving rise to abnormalities such as immunodeficiency or anemia.How HSC homeostasis is maintained has been studied for decades and regulatory factors can be generally categorized into two classes:genetic factors and epigenetic factors.Although genetic factors such as signaling pathways or transcription factors have been well explored,recent studies have emerged the indispensable roles of epigenetic factors.In this review,we have summarized regulatory mechanisms of HSC homeostasis by epigenetic factors,including DNA methylation,histone modification,chromatin remodeling,non-coding RNAs,and RNA modification,which will facilitate applications such as HSC ex vivo expansion and exploration of novel therapeutic approaches for many hematological diseases.
基金National Key Research and Development Program of China,Stem Cell and Translation Research[2018YFA0109300 to P.Q.]National Natural Science Foundation of China[82222003,81870080,91949115,82161138028 to P.Q.+3 种基金31900815 to H.W.]Natural Science Foundation of Zhejiang Province[LR19H080001 to P.Q.]Leading Innovative and Entrepreneur Team Introduction Program of Zhejiang[2020R01006 to P.Q.]Zhejiang Provincial Key Research and Development Program[2021C03043 to H.Y.].We are grateful to W.P.H.(Shandong University)for providing original SARS-CoV-2 protein expressing plasmids.
文摘Dear Editor,COVID-19(Coronavirus Disease 2019)is causing an unprecedented public health crisis.Protein translation is crucial for virus lifecycle.Nucleocapsid(N)protein is among the most abundant SARS-CoV-2 proteins and highly conserved across coronavirus genus.1 However,its function in subverting host translation machinery is still elusive.
基金supported by the National Key Research and Development Program of China(2022YFA1103500 to P.Q.)the National Key Nature Science Foundation of China(82130003 to H.H.)+3 种基金the Key Project of Science and Technology Department of Zhejiang Province(2019C03016 and 2020C03G2013586 to H.H.,Z24H080001 to P.Q.)the National Natural Science Foundation of China(82200248 and 82000194 to H.H.,82222003 and 82161138028 to P.Q.)the Key R&D Program of Zhejiang(2024SSYS0023 to H.H.,2024SSYS0024 to P.Q.,2024SSYS0025 to Y.H.)the Fundamental Research Funds for the Central Universities(226-2024-00007 to P.Q.).
文摘Clinical evidence supports the notion that T cell exhaustion and terminal differentiation pose challenges to the persistence and effectiveness of chimeric antigen receptor-T(CAR-T)cells.MEK1/2 inhibitors(MEKIs),widely used in cancer treatment due to their ability to inhibit aberrant MAPK signaling,have shown potential synergistic effects when combined with immunotherapy.However,the impact and mechanisms of MEKIs on CAR-T cells remain uncertain and controversial.To address this,we conducted a comprehensive investigation to determine whether MEKIs enhance or impair the efficacy of CAR-T cells.Our findings revealed that MEKIs attenuated CAR-T cell exhaustion and terminal differentiation induced by tonic signaling and antigen stimulation,thereby improving CAR-T cell efficacy against hematological and solid tumors.Remarkably,these effects were independent of the specific scFvs and costimulatory domains utilized in CARs.Mechanistically,analysis of bulk and single-cell transcriptional profiles demonstrates that the effect of MEK inhibition was related to diminish anabolic metabolism and downregulation of c-Fos and JunB.Additionally,the overexpression of c-Fos or JunB in CAR-T cells counteracted the effects of MEK inhibition.Furthermore,our Cut-and-Tag assay revealed that MEK inhibition downregulated the JunB-driven gene profiles associated with exhaustion,differentiation,anergy,glycolysis,and apoptosis.In summary,our research unveil the critical role of the MAPK-c-Fos-JunB axis in driving CAR-T cell exhaustion and terminal differentiation.These mechanistic insights significantly broaden the potential application of MEKIs to enhance the effectiveness of CAR-T therapy.