Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 47(20): e130, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31504804

ABSTRACT

Chemical modification of transcripts with 5' caps occurs in all organisms. Here, we report a systems-level mass spectrometry-based technique, CapQuant, for quantitative analysis of an organism's cap epitranscriptome. The method was piloted with 21 canonical caps-m7GpppN, m7GpppNm, GpppN, GpppNm, and m2,2,7GpppG-and 5 'metabolite' caps-NAD, FAD, UDP-Glc, UDP-GlcNAc, and dpCoA. Applying CapQuant to RNA from purified dengue virus, Escherichia coli, yeast, mouse tissues, and human cells, we discovered new cap structures in humans and mice (FAD, UDP-Glc, UDP-GlcNAc, and m7Gpppm6A), cell- and tissue-specific variations in cap methylation, and high proportions of caps lacking 2'-O-methylation (m7Gpppm6A in mammals, m7GpppA in dengue virus). While substantial Dimroth-induced loss of m1A and m1Am arose with specific RNA processing conditions, human lymphoblast cells showed no detectable m1A or m1Am in caps. CapQuant accurately captured the preference for purine nucleotides at eukaryotic transcription start sites and the correlation between metabolite levels and metabolite caps.


Subject(s)
Epigenesis, Genetic , RNA Caps/chemistry , RNA Processing, Post-Transcriptional , Sequence Analysis, RNA/methods , Transcriptome , Animals , Cells, Cultured , Dengue Virus , Female , Humans , Mice , Mice, Inbred C57BL , RNA Caps/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Saccharomyces cerevisiae
2.
Genes Dev ; 27(12): 1378-90, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23788624

ABSTRACT

Although it is known that OCT4-NANOG are required for maintenance of pluripotent cells in vitro, the upstream signals that regulate this circuit during early development in vivo have not been identified. Here we demonstrate, for the first time, signal transducers and activators of transcription 3 (STAT3)-dependent regulation of the OCT4-NANOG circuitry necessary to maintain the pluripotent inner cell mass (ICM), the source of in vitro-derived embryonic stem cells (ESCs). We show that STAT3 is highly expressed in mouse oocytes and becomes phosphorylated and translocates to the nucleus in the four-cell and later stage embryos. Using leukemia inhibitory factor (Lif)-null embryos, we found that STAT3 phosphorylation is dependent on LIF in four-cell stage embryos. In blastocysts, interleukin 6 (IL-6) acts in an autocrine fashion to ensure STAT3 phosphorylation, mediated by janus kinase 1 (JAK1), a LIF- and IL-6-dependent kinase. Using genetically engineered mouse strains to eliminate Stat3 in oocytes and embryos, we firmly establish that STAT3 is essential for maintenance of ICM lineages but not for ICM and trophectoderm formation. Indeed, STAT3 directly binds to the Oct4 and Nanog distal enhancers, modulating their expression to maintain pluripotency of mouse embryonic and induced pluripotent stem cells. These results provide a novel genetic model of cell fate determination operating through STAT3 in the preimplantation embryo and pluripotent stem cells in vivo.


Subject(s)
Blastocyst Inner Cell Mass , Cell Lineage , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins , Octamer Transcription Factor-3 , STAT3 Transcription Factor , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphorylation , Pluripotent Stem Cells/physiology , Protein Binding , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
3.
J Pathol ; 246(1): 89-102, 2018 09.
Article in English | MEDLINE | ID: mdl-29926931

ABSTRACT

Characterising the activated oncogenic signalling that leads to advanced breast cancer is of clinical importance. Here, we showed that SET domain, bifurcated 1 (SETDB1), a histone H3 lysine 9 methyltransferase, is aberrantly expressed and behaves as an oncogenic driver in breast cancer. SETDB1 enhances c-MYC and cyclin D1 expression by promoting the internal ribosome entry site (IRES)-mediated translation of MYC/CCND1 mRNA, resulting in prominent signalling of c-MYC to promote cell cycle progression, and provides a growth/self-renewal advantage to breast cancer cells. The activated c-MYC-BMI1 axis is essential for SETDB1-mediated breast tumourigenesis, because silencing of either c-MYC or BMI1 profoundly impairs the enhanced growth/colony formation conferred by SETDB1. Furthermore, c-MYC directly binds to the SETDB1 promoter region and enhances its transcription, suggesting a positive regulatory interplay between SETDB1 and c-MYC. In this study, we identified SETDB1 as a prominent oncogene and characterised the underlying mechanism whereby SETDB1 drives breast cancer, providing a therapeutic rationale for targeting SETDB1-BMI1 signalling in breast cancer. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Breast Neoplasms/enzymology , Carcinogenesis/metabolism , Polycomb Repressive Complex 1/metabolism , Protein Methyltransferases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Cycle , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , MCF-7 Cells , Mice , Oncogenes , Polycomb Repressive Complex 1/genetics , Protein Methyltransferases/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Transcriptional Activation
4.
J Biol Chem ; 292(35): 14695-14703, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28655767

ABSTRACT

Chemical RNA modifications are central features of epitranscriptomics, highlighted by the discovery of modified ribonucleosides in mRNA and exemplified by the critical roles of RNA modifications in normal physiology and disease. Despite a resurgent interest in these modifications, the biochemistry of 3-methylcytidine (m3C) formation in mammalian RNAs is still poorly understood. However, the recent discovery of trm141 as the second gene responsible for m3C presence in RNA in fission yeast raises the possibility that multiple enzymes are involved in m3C formation in mammals as well. Here, we report the discovery and characterization of three distinct m3C-contributing enzymes in mice and humans. We found that methyltransferase-like (METTL) 2 and 6 contribute m3C in specific tRNAs and that METTL8 only contributes m3C to mRNA. MS analysis revealed that there is an ∼30-40% and ∼10-15% reduction, respectively, in METTL2 and -6 null-mutant cells, of m3C in total tRNA, and primer extension analysis located METTL2-modified m3C at position 32 of tRNAThr isoacceptors and tRNAArg(CCU) We also noted that METTL6 interacts with seryl-tRNA synthetase in an RNA-dependent manner, suggesting a role for METTL6 in modifying serine tRNA isoacceptors. METTL8, however, modified only mRNA, as determined by biochemical and genetic analyses in Mettl8 null-mutant mice and two human METTL8 mutant cell lines. Our findings provide the first evidence of the existence of m3C modification in mRNA, and the discovery of METTL8 as an mRNA m3C writer enzyme opens the door to future studies of other m3C epitranscriptomic reader and eraser functions.


Subject(s)
Cytidine/analogs & derivatives , Liver/metabolism , Methyltransferases/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Animals , Cell Line , Cytidine/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/enzymology , Methylation , Methyltransferases/antagonists & inhibitors , Methyltransferases/chemistry , Methyltransferases/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Mutation , RNA Interference , RNA, Transfer, Arg/metabolism , RNA, Transfer, Ser/metabolism , RNA, Transfer, Thr/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/metabolism , Substrate Specificity
5.
Circulation ; 133(1): 48-61, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26628621

ABSTRACT

BACKGROUND: ß-Adrenergic receptors (ßARs) play paradoxical roles in the heart. On one hand, ßARs augment cardiac performance to fulfill the physiological demands, but on the other hand, prolonged activations of ßARs exert deleterious effects that result in heart failure. The signal transducer and activator of transcription 3 (STAT3) plays a dynamic role in integrating multiple cytokine signaling pathways in a number of tissues. Altered activation of STAT3 has been observed in failing hearts in both human patients and animal models. Our objective is to determine the potential regulatory roles of STAT3 in cardiac ßAR-mediated signaling and function. METHODS AND RESULTS: We observed that STAT3 can be directly activated in cardiomyocytes by ß-adrenergic agonists. To follow up this finding, we analyzed ßAR function in cardiomyocyte-restricted STAT3 knockouts and discovered that the conditional loss of STAT3 in cardiomyocytes markedly reduced the cardiac contractile response to acute ßAR stimulation, and caused disengagement of calcium coupling and muscle contraction. Under chronic ß-adrenergic stimulation, Stat3cKO hearts exhibited pronounced cardiomyocyte hypertrophy, cell death, and subsequent cardiac fibrosis. Biochemical and genetic data supported that Gαs and Src kinases are required for ßAR-mediated activation of STAT3. Finally, we demonstrated that STAT3 transcriptionally regulates several key components of ßAR pathway, including ß1AR, protein kinase A, and T-type Ca(2+) channels. CONCLUSIONS: Our data demonstrate for the first time that STAT3 has a fundamental role in ßAR signaling and functions in the heart. STAT3 serves as a critical transcriptional regulator for ßAR-mediated cardiac stress adaption, pathological remodeling, and heart failure.


Subject(s)
Heart/physiology , Receptors, Adrenergic, beta/physiology , STAT3 Transcription Factor/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Cell Line , Heart/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Organ Culture Techniques
6.
Dev Biol ; 400(1): 1-9, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25536399

ABSTRACT

In the early mouse embryo, a specialized population of extraembryonic visceral endoderm (VE) cells called the distal VE (DVE) arises at the tip of the egg cylinder stage embryo and then asymmetrically migrates to the prospective anterior, recruiting additional distal cells. Upon migration these cells, called the anterior VE (AVE), establish the anterior posterior (AP) axis by restricting gastrulation-inducing signals to the opposite pole. The Nodal-signaling pathway has been shown to have a critical role in the generation and migration of the DVE/AVE. The Nodal gene is expressed in both the VE and in the pluripotent epiblast, which gives rise to the germ layers. Previous findings have provided conflicting evidence as to the relative importance of Nodal signaling from the epiblast vs. VE for AP patterning. Here we show that conditional mutagenesis of the Nodal gene specifically within the VE leads to reduced Nodal expression levels in the epiblast and incomplete or failed DVE/AVE migration. These results support a required role for VE Nodal to maintain normal levels of expression in the epiblast, and suggest signaling from both VE and epiblast is important for DVE/AVE migration.


Subject(s)
Body Patterning/physiology , Cell Movement/physiology , Endoderm/physiology , Gene Expression Regulation, Developmental/physiology , Germ Layers/metabolism , Nodal Protein/metabolism , Signal Transduction/physiology , Animals , Endoderm/cytology , Galactosides , Genes, Reporter/genetics , In Situ Hybridization , Indoles , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mutagenesis , Nodal Protein/genetics
7.
Blood ; 120(13): 2589-99, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22665934

ABSTRACT

Nuclear transcription factor Stat3 is important for proper regulation of hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) proliferation, survival, and cytokine signaling responses. A new, noncanonical role for Stat3 in mitochondrial function has been discovered recently. However, there is little information on the role(s) of mitochondrial Stat3 in HSC/HPC function, especially potential effects of Stat3/mitochondrial dysregulation in human diseases. We investigated hematopoietic cell-targeted deletion of the STAT3 gene in HSCs/HPCs with a focus on mitochondrial function. We found that STAT3(-/-) mice, which have a very shortened lifespan, dysfunctional/dysregulated mitochondrial function and excessive reactive oxygen species production in HSCs/HPCs that coincides with pronounced defects in function. These animals have a blood phenotype with similarities to premature aging and to human diseases of myelodysplastic syndrome and myeloproliferative neoplasms such as erythroid dysplasia, anemia, excessive myeloproliferation, and lymphomyeloid ratio shifts. We show herein that the lifespan of STAT3(-/-) animals is lengthened by treatment with a reactive oxygen species scavenger, which lessened the severity of the blood phenotype. These data suggest a need for more detailed studies of role(s) of Stat3 in HSC/HPC mitochondrial function in human diseases and raise the idea that mitochondrial Stat3 could be used as a potential therapeutic target.


Subject(s)
Aging/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/physiology , Acetylcysteine/pharmacology , Anemia , Animals , Bone Marrow/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Erythroid Cells/cytology , Erythroid Cells/drug effects , Female , Free Radical Scavengers/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption/drug effects , Phenotype , Sequence Deletion
8.
ACS Appl Mater Interfaces ; 16(15): 18959-18970, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38569111

ABSTRACT

Fe-based Prussian blue (Fe-PB) analogues have emerged as promising cathode materials for sodium-ion batteries, owing to their cost-effectiveness, high theoretical capacity, and environmental friendliness. However, their practical application is hindered by [Fe(CN)6] defects, negatively impacting capacity and cycle stability. This work reports a hollow layered Fe-PB composite material using 1,3,5-benzenetricarboxylic acid (BTA) as a chelating and etching agent by the hydrothermal method. Compared to benzoic acid, our approach significantly reduces defects and enhances the yield of Fe-PB. Notably, the hollow layered structure shortens the diffusion path of sodium ions, enhances the activity of low-spin Fe in the Fe-PB lattice, and mitigates volume changes during Na-ion insertion/extraction into/from Fe-PB. As a sodium-ion battery cathode, this hollow layered Fe-PB exhibits an impressive initial capacity of 95.9 mAh g-1 at a high current density of 1 A g-1. Even after 500 cycles, it still maintains a considerable discharge capacity of 73.1 mAh g-1, showing a significantly lower capacity decay rate (0.048%) compared to the control sample (0.089%). Moreover, the full cell with BTA-PB-1.6 as the cathode and HC as the anode provides a considerable energy density of 312.2 Wh kg-1 at a power density of 291.0 W kg-1. This research not only enhances the Na storage performance of Fe-PB but also increases the yield of products obtained by hydrothermal methods, providing some technical reference for the production of PB materials using the low-yield hydrothermal method.

9.
Nat Protoc ; 18(9): 2671-2698, 2023 09.
Article in English | MEDLINE | ID: mdl-37567932

ABSTRACT

Chemical modifications of transcripts with a 5' cap occur in all organisms and function in many aspects of RNA metabolism. To facilitate analysis of RNA caps, we developed a systems-level mass spectrometry-based technique, CapQuant, for accurate and sensitive quantification of the cap epitranscriptome. The protocol includes the addition of stable isotope-labeled cap nucleotides (CNs) to RNA, enzymatic hydrolysis of endogenous RNA to release CNs, and off-line enrichment of CNs by ion-pairing high-pressure liquid chromatography, followed by a 17 min chromatography-coupled tandem quadrupole mass spectrometry run for the identification and quantification of individual CNs. The total time required for the protocol can be up to 7 d. In this approach, 26 CNs can be quantified in eukaryotic poly(A)-tailed RNA, bacterial total RNA and viral RNA. This protocol can be modified to analyze other types of RNA and RNA from in vitro sources. CapQuant stands out from other methods in terms of superior specificity, sensitivity and accuracy, and it is not limited to individual caps nor does it require radiolabeling. Thanks to its unique capability of accurately and sensitively quantifying RNA caps on a systems level, CapQuant can reveal both the RNA cap landscape and the transcription start site distribution of capped RNA in a broad range of settings.


Subject(s)
RNA Caps , Tandem Mass Spectrometry , RNA Caps/genetics , RNA, Messenger/genetics , Chromatography, High Pressure Liquid , RNA, Viral/genetics , RNA, Bacterial
10.
Nature ; 441(7096): 1015-9, 2006 Jun 22.
Article in English | MEDLINE | ID: mdl-16791201

ABSTRACT

SMAD4 (MAD homologue 4 (Drosophila)), also known as DPC4 (deleted in pancreatic cancer), is a tumour suppressor gene that encodes a central mediator of transforming growth factor-beta signalling. Germline mutations in SMAD4 are found in over 50% of patients with familial juvenile polyposis, an autosomal dominant disorder characterized by predisposition to hamartomatous polyps and gastrointestinal cancer. Dense inflammatory cell infiltrates underlay grossly normal appearing, non-polypoid colonic and gastric mucosa of patients with familial juvenile polyposis. This prominent stromal component suggests that loss of SMAD4-dependent signalling in cells within the epithelial microenvironment has an important role in the evolution of intestinal tumorigenesis in this syndrome. Here we show that selective loss of Smad4-dependent signalling in T cells leads to spontaneous epithelial cancers throughout the gastrointestinal tract in mice, whereas epithelial-specific deletion of the Smad4 gene does not. Tumours arising within the colon, rectum, duodenum, stomach and oral cavity are stroma-rich with dense plasma cell infiltrates. Smad4(-/-) T cells produce abundant T(H)2-type cytokines including interleukin (IL)-5, IL-6 and IL-13, known mediators of plasma cell and stromal expansion. The results support the concept that cancer, as an outcome, reflects the loss of the normal communication between the cellular constituents of a given organ, and indicate that Smad4-deficient T cells ultimately send the wrong message to their stromal and epithelial neighbours.


Subject(s)
Gastrointestinal Neoplasms/immunology , Signal Transduction , Smad4 Protein/metabolism , T-Lymphocytes/metabolism , Adenomatous Polyposis Coli/etiology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Cell Communication , Cytokines/biosynthesis , Disease Models, Animal , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gene Deletion , Gene Targeting , Mice , Mice, Inbred C57BL , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Smad4 Protein/genetics
11.
ACS Appl Mater Interfaces ; 14(38): 43308-43318, 2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36107796

ABSTRACT

The Fe-based Prussian blue (Fe-PB) composite is considered as one of the most potential cathode materials for sodium-ion batteries because of its abundant iron resources and high theoretical capacity. However, the crystal water and vacancy in the Fe-PB structure will lead to poor capacity and cycle stability. In this work, a Cu-modified Fe-PB composite (FeCu-PB@CuO) is successfully prepared through regulating the Fe-PB structure by Cu doping and engineering the surface by CuO coating. The density functional theory calculation results confirm that Cu preferentially replaces FeHS in the Fe-PB lattice and Cu doping reduces the bandgap. Our experiment results reveal that CuO coating can provide more active sites, inhibit side reactions, and potentially enhance the activity of FeHS. Due to the synergistic effect of Cu doping and CuO coating, FeCu-PB@CuO has a considerable initial discharge capacity of 123.5 mAh g-1 at 0.1 A g-1. In particular, at 2 A g-1, it delivers an impressive initial capacity of 84.3 mAh g-1, and the capacity decreasing rate of each cycle is only 0.02% over 1500 cycles. Therefore, the synergistic modification strategy of metal ion doping and metal oxide coating has tremendous application potential and can be extended to other electrode materials.

12.
ACS Appl Mater Interfaces ; 14(4): 5506-5513, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35072463

ABSTRACT

Iron-based Prussian blue (FeHCF) has great application potential in the large-scale production of sodium-ion (Na+) batteries because of its high theoretical capacity and abundant Fe ore resources. However, the Fe(CN)6 vacancies and crystal water seriously affect the electrochemical performance. Herein, a Cu-doped FeHCF (Cu-FeHCF) cathode material is successfully prepared directly by a coprecipitation method. After Cu doping, the monoclinic structure and the quasi-cubic morphology are retained, but the electrochemical performance is significantly improved. In addition to few Fe(CN)6 vacancies and low crystal water, the improved performance is also related to the enhanced electrochemical activity of low-spin Fe and the stabilizing effect of Cu on the crystal structure. Moreover, Cu doping also controls the side reaction to a certain extent. As a result, after Cu doping, the initial discharge capacity is enhanced from 107.9 to 127.4 mA h g-1 at 100 mA g-1, especially the capacities contributed by low-spin Fe increase from 30.0, 21.7, and 16.7 mA h g-1 to 48.8, 45.4, and 43.7 mA h g-1 for the first three cycles, respectively. Even at 2 A g-1, Cu-FeHCF still has a promising initial capacity of 82.3 mA h g-1 and only a 0.047% capacity decay rate for each cycle over 500 cycles. Therefore, Cu-FeHCF shows excellent application potential in the field of Na+ energy storage batteries.

13.
Sci China Life Sci ; 65(9): 1824-1839, 2022 09.
Article in English | MEDLINE | ID: mdl-35508790

ABSTRACT

Signal transducers and activators of transcription 5 (Stat5) is known to engage in regulating the differentiation and effector function of various subsets of T helper cells. However, how Stat5 regulates the antitumor activity of tumor-infiltrating CD4+ T cells is largely unknown. Here, we showed that mice with specific deletion of Stat5 in CD4+ T cells were less susceptible to developing subcutaneous and lung metastatic B16 melanoma with CD4+ tumor-infiltrating lymphocytes (TILs) remolding. Especially, we confirmed that Stat5-deficient CD4+ naïve T cells were prone to polarization of two subtypes of Th17 cells: IFN-γ+ and IFN-γ- Th17 cells, which exhibited increased anti-melanoma activity through enhanced activation of Notch1 pathway compared with wild type Th17 cells. Our study therefore revealed a novel function of Stat5 in regulating tumor-specific Th17 cell differentiation and function in melanoma. This study also provided a new possibility for targeting Stat5 and other Th17-associated pathways to develop novel immunotherapies for melanoma patients.


Subject(s)
Melanoma , T-Lymphocytes, Helper-Inducer , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Lymphocytes, Tumor-Infiltrating , Melanoma/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
14.
Hepatology ; 51(4): 1354-62, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20041412

ABSTRACT

UNLABELLED: Liver regeneration triggered by two-thirds partial hepatectomy is accompanied by elevated hepatic levels of endotoxin, which contributes to the regenerative process, but liver inflammation and apoptosis remain paradoxically limited. Here, we show that signal transducer and activator of transcription 3 (STAT3), an important anti-inflammatory signal, is activated in myeloid cells after partial hepatectomy and its conditional deletion results in an enhanced inflammatory response. Surprisingly, this is accompanied by an improved rather than impaired regenerative response with increased hepatic STAT3 activation, which may contribute to the enhanced liver regeneration. Indeed, conditional deletion of STAT3 in both hepatocytes and myeloid cells results in elevated activation of STAT1 and apoptosis of hepatocytes, and a dramatic reduction in survival after partial hepatectomy, whereas additional global deletion of STAT1 protects against these effects. CONCLUSION: An interplay of myeloid and hepatic STAT3 signaling is essential to prevent liver failure during liver regeneration through tempering a strong innate inflammatory response mediated by STAT1 signaling.


Subject(s)
Hepatocytes/physiology , Immunity, Innate , Liver Regeneration , Myeloid Cells/physiology , STAT3 Transcription Factor/physiology , Animals , Hepatectomy , Interferon-gamma/blood , Male , Mice , STAT1 Transcription Factor/physiology
15.
Hepatology ; 51(5): 1724-34, 2010 May.
Article in English | MEDLINE | ID: mdl-20196117

ABSTRACT

UNLABELLED: Liver injury is associated with inflammation, which is generally believed to accelerate the progression of liver diseases; however, clinical data show that inflammation does not always correlate with hepatocelluar damage in some patients. Investigating the cellular mechanisms underlying these events using an experimental animal model, we show that inflammation may attenuate liver necrosis induced by carbon tetrachloride (CCl(4)) in myeloid-specific signal transducer and activator of transcription 3 (STAT3) knockout mice. As an important anti-inflammatory signal, conditional deletion of STAT3 in myeloid cells results in markedly enhanced liver inflammation after CCl(4) injection. However, these effects are also accompanied by reduced liver necrosis, correlating with elevated serum interleukin-6 (IL-6) and hepatic STAT3 activation. An additional deletion of STAT3 in hepatocytes in myeloid-specific STAT3 knockout mice restored hepatic necrosis but decreased liver inflammation. CONCLUSION: Inflammation-mediated STAT3 activation attenuates hepatocellular injury induced by CCl(4) in myeloid-specific STAT3 knockout mice, suggesting that inflammation associated with a predominance of hepatoprotective cytokines that activate hepatic STAT3 may reduce rather than accelerate hepatocellular damage in patients with chronic liver diseases.


Subject(s)
Carbon Tetrachloride Poisoning/pathology , Inflammation/pathology , Liver/pathology , STAT3 Transcription Factor/genetics , Animals , Hepatocytes/metabolism , Inflammation/chemically induced , Liver/metabolism , Male , Mice , Mice, Knockout , Necrosis/chemically induced , Oxidative Stress , STAT3 Transcription Factor/physiology
16.
Blood ; 114(2): 394-403, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19411632

ABSTRACT

Internal tandem duplication mutations in the Flt3 tyrosine kinase gene (ITD-Flt3) and overexpression of Survivin are frequently found in patients with acute myeloid leukemia (AML). We investigated whether Survivin mediates the enhanced survival of primary hematopoietic progenitor cells (HPCs) resulting from ITD-Flt3 signaling. Ectopic ITD-Flt3 mutants increased Survivin expression in Ba/F3 cells downstream of PI3-kinase/Akt. Treatment of ITD-Flt3(+) human MV4-11 leukemia cells with the ITD-Flt3 inhibitor SU5416 reduced Survivin expression and inhibited cell proliferation. ITD-Flt3 dramatically increased the number of primary mouse marrow c-kit(+), Sca-1(+), Lin(Neg) cells and colony-forming unit granulocyte-macrophages (CFU-GMs) able to proliferate in the absence of growth factors, whereas Survivin deletion significantly reduced growth factor-independent proliferation and increased apoptosis, which was further accentuated by SU5416. Ectopic ITD-Flt3 reduced differentiation of Lin(Neg) marrow cells cultured with granulocyte-macrophage colony-stimulating factor (GM-CSF) plus stem cell factor, which was partially blocked by Survivin deletion. In addition, Survivin deletion decreased secondary colony formation induced by ITD-Flt3. Dominant-negative (dn)-Survivin delayed development of acute leukemia in mice that received a transplant of Ba/F3 cells expressing ITD-Flt3. These results suggest that Survivin regulates expansion of ITD-Flt3-transformed HPCs with self-renewal capability and development of ITD-Flt3(+) acute leukemia and that antagonizing Survivin may provide therapeutic benefit for patients with acute leukemia expressing ITD-Flt3.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Leukemia/metabolism , Microtubule-Associated Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Acute Disease , Animals , Caspase 3/metabolism , Cell Proliferation , Enzyme Activation , Gene Deletion , Humans , Inhibitor of Apoptosis Proteins , Leukemia/genetics , Leukemia/pathology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Mutation/genetics , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins , Survivin , Tumor Cells, Cultured , Up-Regulation , fms-Like Tyrosine Kinase 3/genetics
17.
J Exp Med ; 198(10): 1517-25, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14623907

ABSTRACT

Endothelial cells (ECs) are believed to be an important component in the protection from lipopolysaccharide (LPS)-induced endotoxic shock. However, the cellular and molecular mechanism is not well defined. Here, we report that signal transducer and activator of transcription (STAT) 3 is an essential regulator of the antiinflammatory function of ECs in systemic immunity. Because STAT3 deficiency results in early embryonic lethality, we have generated mice with a conditional STAT3 deletion in endothelium (STAT3E-/-). STAT3E-/- mice are healthy and fertile, and isolated ECs initiate normal tube formation in vitro. Conditional endothelial but not organ-specific (i.e., hepatocyte or cardiomyocyte) STAT3 knockout mice show an increased susceptibility to lethality after LPS challenge. The LPS response in STAT3E-/- mice shows exaggerated inflammation and leukocyte infiltration in multiple organs combined with elevated activity of serum alanine aminotransferase and aspartate aminotransferase, indicating organ damage. Concomitantly, proinflammatory cytokines are produced at an exaggerated level and for a prolonged period. This defect cannot be explained by lack of antiinflammatory cytokines, such as interleukin 10 and transforming growth factor beta. Instead, we have shown that a soluble activity derived from endothelia and dependent on STAT3 is critical for suppression of interferon gamma. These data define STAT3 signaling within endothelia as a critical antiinflammatory mediator and provide new insight to the protective function of ECs in inflammation.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelium/metabolism , Endotoxins/metabolism , Inflammation/metabolism , Trans-Activators/metabolism , Animals , Cytokines/metabolism , DNA-Binding Proteins/genetics , Integrases/genetics , Integrases/metabolism , Interferon-gamma/antagonists & inhibitors , Mice , Mice, Transgenic , Mutation , STAT3 Transcription Factor , Trans-Activators/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Gastroenterology ; 137(6): 2125-35.e1-2, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19686746

ABSTRACT

BACKGROUND & AIMS: T cell-mediated hepatitis is a leading cause of acute liver failure; there is no effective treatment, and the mechanisms underlying its pathogenesis are obscure. The aim of this study was to investigate the immune cell-signaling pathways involved-specifically the role of signal transducer and activator of transcription 3 (STAT3)-in T cell-mediated hepatitis in mice. METHODS: T cell-mediated hepatitis was induced in mice by injection of concanavalin A (Con A). Mice with myeloid cell-specific and T-cell-specific deletion of STAT3 were generated. RESULTS: STAT3 was activated in myeloid and T cells following Con A injection. Deletion of STAT3 specifically from myeloid cells exacerbated T-cell hepatitis and induced STAT1-dependent production of a T helper cell (Th)1 cytokine (interferon [IFN]-gamma) and to a lesser extent of Th17 cytokines (interleukin [IL]-17 and IL-22) in a STAT1-independent manner. In contrast, deletion of STAT3 in T cells reduced T cell-mediated hepatitis and IL-17 production. Furthermore, deletion of IFN-gamma completely abolished Con A-induced T-cell hepatitis, whereas deletion of IL-17 slightly but significantly reduced such injury. In vitro experiments indicated that IL-17 promoted liver inflammation but inhibited hepatocyte apoptosis. CONCLUSIONS: Myeloid STAT3 activation inhibits T cell-mediated hepatitis via suppression of a Th1 cytokine (IFN-gamma) in a STAT1-dependent manner, whereas STAT3 activation in T cells promotes T-cell hepatitis to a lesser extent, via induction of IL-17. Therefore, activation of STAT3 in myeloid cells could be a novel therapeutic strategy for patients with T-cell hepatitis.


Subject(s)
Hepatitis, Autoimmune/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Liver/immunology , Myeloid Cells/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction , Th1 Cells/immunology , Animals , Apoptosis , Cells, Cultured , Concanavalin A , Disease Models, Animal , Hepatitis, Autoimmune/pathology , Hepatitis, Autoimmune/prevention & control , Immunity, Innate , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/metabolism , Liver/pathology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Time Factors , Interleukin-22
19.
Blood ; 112(5): 1832-43, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18577706

ABSTRACT

The clinical outcomes of dendritic cell (DC)-based immunotherapy remain disappointing, with DCs often displaying a tenuous capacity to complete maturation and DC1 polarization in the tumor host. Surprisingly, we observed that the capacity for successful DC1 polarization, including robust IL12p70 production, could be regulated by STAT-dependent events even prior to DC differentiation. Exposure of CD34(pos) cells to single-agent granulocyte-macrophage colony-stimulating factor (GMCSF) induced multilineage, STAT5-dependent differentiation, including DCs that failed to mature in the absence of further exogenous signals. In contrast, Flt3L induced nearly global differentiation of CD34(pos) cells into spontaneously maturing DCs. IL-6 synergized with Flt3L to produce explosive, STAT3-dependent proliferation of phenotypically undifferentiated cells that nevertheless functioned as committed DC1 precursors. Such precursors not only resisted many tumor-associated immunosuppressants, but also responded to tumor contact or TGFbeta with facilitated DC maturation and IL12p70 production, and displayed a superior capacity to reverse tumor-induced T-cell tolerance. GMCSF preempted Flt3L or Flt3L plus IL-6 licensing by blocking STAT3 activation and promoting STAT5-dependent differentiation. Paradoxically, following overt DC differentiation, STAT5 enhanced whereas STAT3 inhibited DC1 polarization. Therefore, nonoverlapping, sequential activation of STAT3 and STAT5, achievable by sequenced exposure to Flt3L plus IL-6, then GMCSF, selects for multilog expansion, programming, and DC1 polarization of tumor-competent DCs from CD34(pos) cells.


Subject(s)
Antigens, CD34/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Differentiation/physiology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunotherapy, Adoptive , Interleukin-6/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Recombinant Proteins , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics
20.
Alcohol Clin Exp Res ; 34(4): 719-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20102572

ABSTRACT

BACKGROUND: It is generally believed that the hepatoprotective effect of interleukin-6 (IL-6) is mediated via activation of signal transducer and activator of transcription 3 (STAT3) in hepatocytes. IL-6-deficient mice are more susceptible to alcohol-induced hepatocyte apoptosis and steatosis and elevation of serum alanine transaminase (ALT); however, whereas hepatocyte-specific STAT3 knockout mice are more susceptible to alcohol-induced hepatic steatosis, they have similar hepatocyte apoptosis and serum ALT after alcohol feeding compared with wild-type mice. This suggests that the hepatoprotective effect of IL-6 in alcoholic liver injury may be mediated via activation of STAT3-independent signals in hepatocytes, activation of STAT3 in nonparenchymal cells, or both. We have previously shown that IL-6 also activates STAT3 in sinusoidal endothelial cells (SECs). Thus, the purpose of this study was to investigate whether STAT3 in endothelial cells also plays a protective role in alcoholic liver injury. METHODS: Wild-type and endothelial cell-specific STAT3 knockout (STAT3(E-/-)) mice were pair-fed and fed ethanol containing diet for 4 weeks. Liver injury and inflammation were determined. RESULTS: Feeding mice with ethanol-containing diet for 4 weeks induced greater hepatic injury (elevation of serum ALT) and liver weight in STAT3(E-/-) mice than wild-type control groups. In addition, ethanol-fed STAT3(E-/-) mice displayed greater hepatic inflammation and substantially elevated serum and hepatic levels of IL-6 and TNF-alpha compared with wild-type mice. Furthermore, ethanol-fed STAT3(E-/-) mice displayed a greater abundance of apoptotic SECs and higher levels of serum hyaluronic acid than wild-type controls. CONCLUSIONS: These data suggest that endothelial cell STAT3 plays important dual functions of attenuating hepatic inflammation and SEC death during alcoholic liver injury.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Endothelium, Vascular/metabolism , Hepatitis, Alcoholic/metabolism , Inflammation Mediators/physiology , STAT3 Transcription Factor/physiology , Animals , Apoptosis/drug effects , Endothelium, Vascular/pathology , Ethanol/toxicity , Hepatitis, Alcoholic/pathology , Hepatitis, Alcoholic/prevention & control , Inflammation Mediators/metabolism , Interleukin-6/deficiency , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/prevention & control , Male , Mice , Mice, Knockout , Mice, Transgenic , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL