Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Nat Immunol ; 18(7): 733-743, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28481329

ABSTRACT

The transcription regulator YAP controls organ size by regulating cell growth, proliferation and apoptosis. However, whether YAP has a role in innate antiviral immunity is largely unknown. Here we found that YAP negatively regulated an antiviral immune response. YAP deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in vivo. YAP blocked dimerization of the transcription factor IRF3 and impeded translocation of IRF3 to the nucleus after viral infection. Notably, virus-activated kinase IKKɛ phosphorylated YAP at Ser403 and thereby triggered degradation of YAP in lysosomes and, consequently, relief of YAP-mediated inhibition of the cellular antiviral response. These findings not only establish YAP as a modulator of the activation of IRF3 but also identify a previously unknown regulatory mechanism independent of the kinases Hippo and LATS via which YAP is controlled by the innate immune pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Fibroblasts/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/immunology , Lysosomes/metabolism , Macrophages/immunology , Phosphoproteins/immunology , Rhabdoviridae Infections/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CRISPR-Cas Systems , Cell Cycle Proteins , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Fluorescent Antibody Technique , Gene Editing , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Lung/immunology , Lung/pathology , Mice , Microscopy, Confocal , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdoviridae Infections/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Vesiculovirus , Viral Load , YAP-Signaling Proteins
3.
Cell ; 151(3): 576-89, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23101626

ABSTRACT

Embryonic stem cell (ESC) pluripotency requires bivalent epigenetic modifications of key developmental genes regulated by various transcription factors and chromatin-modifying enzymes. How these factors coordinate with one another to maintain the bivalent chromatin state so that ESCs can undergo rapid self-renewal while retaining pluripotency is poorly understood. We report that Utf1, a target of Oct4 and Sox2, is a bivalent chromatin component that buffers poised states of bivalent genes. By limiting PRC2 loading and histone 3 lysine-27 trimethylation, Utf1 sets proper activation thresholds for bivalent genes. It also promotes nuclear tagging of messenger RNAs (mRNAs) transcribed from insufficiently silenced bivalent genes for cytoplasmic degradation through mRNA decapping. These opposing functions of Utf1 promote coordinated differentiation. The mRNA degradation function also ensures rapid cell proliferation by blocking the Myc-Arf feedback control. Thus, Utf1 couples the core pluripotency factors with Myc and PRC2 networks to promote the pluripotency and proliferation of ESCs.


Subject(s)
Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Pluripotent Stem Cells/metabolism , RNA, Messenger/metabolism , Trans-Activators/metabolism , ADP-Ribosylation Factors/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Humans , Pluripotent Stem Cells/cytology , Proto-Oncogene Proteins c-myc/metabolism
4.
Nucleic Acids Res ; 51(9): e50, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36938898

ABSTRACT

Genetic screening based on the clustered regularly interspaced palindromic repeat (CRISPR) system has been indicated to be a powerful tool for identifying regulatory genes or cis-elements. However, when applying CRISPR screens to pinpoint functional elements at particular loci, a large number of guide RNA (gRNA) spacers may be required to achieve saturated coverage. Here, we present a controlled template-dependent elongation (CTDE) method relying on reversible terminators to synthesize gRNA libraries with genomic regions of interest. By applying this approach to H3K4me3 chromatin immunoprecipitation (ChIP)-derived DNA of mammalian cells, mega-sized gRNA libraries were synthesized in a tissue-specific manner, with which we conducted screening experiments to annotate essential sites for cell proliferation. Additionally, we confirmed that an essential site within the intron of LINC00339 regulates its own mRNA and that LINC00339 is a novel regulator of the cell cycle that maintains HepG2 proliferation. The CTDE method has the potential to be automated with high efficiency at low cost, and will be widely used to identify functional elements in mammalian genomes.


Subject(s)
Gene Library , Genome , Histones , Mammals , RNA, Guide, CRISPR-Cas Systems , Animals , Humans , Cell Proliferation , Chromatin Immunoprecipitation , Clustered Regularly Interspaced Short Palindromic Repeats , CRISPR-Cas Systems , DNA/genetics , Genome/genetics , Genomics , Hep G2 Cells , Histones/genetics , Mammals/genetics , Organ Specificity , Cell Cycle/genetics , Automation
5.
Nutr Cancer ; 76(2): 215-225, 2024.
Article in English | MEDLINE | ID: mdl-38044546

ABSTRACT

Colon cancer (COAD) is a prevalent gastrointestinal tumor, composed of a few cancer stem cells (CSCs). High expression of RNF183 drives colorectal cancer metastasis, but its role in COAD cell stemness is still unclear. Bioinformatics analyzed expression and enriched pathway of RNF183 in COAD tissue. IHC analyzed RNF183 protein expression in tumor tissue. CD133 + CD44+ CSCs were sorted by flow cytometry, and RNF183 expression in COAD cells or CSCs was detected by qPCR, western blot and immunofluorescence. CCK-8 assay assessed cell viability, and sphere formation assay tested cell sphere-forming ability. Western blot measured protein expression of stem cell markers. qPCR assayed expression of fatty acid oxidation genes. The ability of fatty acid oxidation was analyzed by detecting fatty acid metabolism. RNF183 was highly expressed in COAD and CD133 + CD44+ CSCs, and was enriched in fatty acid metabolism pathway. RNF183 expression was positively correlated with enzymes involved in fatty acid oxidation. RNF183 could promote COAD stemness and fatty acid oxidation. Rescue experiments showed that Orlistat (a fatty acid oxidation inhibitor) reversed stimulative impact of RNF183 overexpression on COAD stemness. RNF183 promoted COAD stemness by affecting fatty acid oxidation, which may be a new therapeutic target for inhibiting COAD development.


Subject(s)
Colonic Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/pathology , Cell Movement , Fatty Acids/metabolism , Neoplastic Stem Cells/pathology , Gene Expression Regulation, Neoplastic , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
J Biol Chem ; 293(14): 5160-5171, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29475944

ABSTRACT

The liver is a major organ in lipid metabolism, and its malfunction leads to various diseases. Nonalcoholic fatty liver disease, the most common chronic liver disorder in developed countries, is characterized by the abnormal retention of excess lipid within hepatocytes and predisposes individuals to liver cancer. We previously reported that the levels of Lissencephaly 1 (LIS1, also known as PAFAH1B1) are down-regulated in human hepatocellular carcinoma. Following up on this observation, we found that genetic deletion of Lis1 in the mouse liver increases lipid accumulation and inflammation in this organ. Further analysis revealed that loss of Lis1 triggers endoplasmic reticulum (ER) stress and reduces triglyceride secretion. Attenuation of ER stress by addition of tauroursodeoxycholic acid (TUDCA) diminished lipid accumulation in the Lis1-deficient hepatocytes. Moreover, the Golgi stacks were disorganized in Lis1-deficient liver cells. Of note, the Lis1 liver-knockout mice exhibited increased hepatocyte ploidy and accelerated development of liver cancer after exposure to the liver carcinogen diethylnitrosamine (DEN). Taken together, these findings suggest that reduced Lis1 levels can spur the development of liver diseases from steatosis to liver cancer and provide a useful model for delineating the molecular pathways that lead to these diseases.


Subject(s)
Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Classical Lissencephalies and Subcortical Band Heterotopias/metabolism , Fatty Liver/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Hepatocytes/metabolism , Lipid Metabolism , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/metabolism
7.
BMC Genomics ; 19(1): 50, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29334893

ABSTRACT

BACKGROUND: Highly polymorphic human leukocyte antigen (HLA) genes are responsible for fine-tuning the adaptive immune system. High-resolution HLA typing is important for the treatment of autoimmune and infectious diseases. Additionally, it is routinely performed for identifying matched donors in transplantation medicine. Although many HLA typing approaches have been developed, the complexity, low-efficiency and high-cost of current HLA-typing assays limit their application in population-based high-throughput HLA typing for donors, which is required for creating large-scale databases for transplantation and precision medicine. RESULTS: Here, we present a cost-efficient Saturated Tiling Capture Sequencing (STC-Seq) approach to capturing 14 HLA class I and II genes. The highly efficient capture (an approximately 23,000-fold enrichment) of these genes allows for simplified allele calling. Tests on five genes (HLA-A/B/C/DRB1/DQB1) from 31 human samples and 351 datasets using STC-Seq showed results that were 98% consistent with the known two sets of digitals (field1 and field2) genotypes. Additionally, STC can capture genomic DNA fragments longer than 3 kb from HLA loci, making the library compatible with the third-generation sequencing. CONCLUSIONS: STC-Seq is a highly accurate and cost-efficient method for HLA typing which can be used to facilitate the establishment of population-based HLA databases for the precision and transplantation medicine.


Subject(s)
High-Throughput Nucleotide Sequencing , Histocompatibility Testing/methods , Sequence Analysis, DNA , HLA Antigens/genetics , Humans
8.
Mol Cell ; 31(1): 143-51, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18614053

ABSTRACT

Sister chromatid cohesion is normally established in S phase in a process that depends on the cohesion establishment factor Eco1, a conserved acetyltransferase. However, due to the lack of known in vivo substrates, how Eco1 regulates cohesion is not understood. Here we report that yeast Eco1 and its human ortholog, ESCO1, both acetylate Smc3, a component of the cohesin complex that physically holds the sister chromatid together, at two conserved lysine residues. Mutating these lysine residues to a nonacetylatable form leads to increased loss of sister chromatid cohesion and genome instability in both yeast and human. In addition, we clarified that the acetyltransferase activity of Eco1 is essential for its function. Our study thus identified a molecular target for the acetyltransferase Eco1 and revealed that Smc3 acetylation is a conserved mechanism in regulating sister chromatid cohesion.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , S Phase , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Sister Chromatid Exchange , Acetylation , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Line , Cell Proliferation , Cell Survival , Chondroitin Sulfate Proteoglycans/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Genomic Instability , Humans , Lysine/metabolism , Molecular Sequence Data , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Substrate Specificity
9.
J Neurosci Res ; 90(2): 422-34, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22002503

ABSTRACT

In vitro models are important tools for studying the mechanisms that govern neuronal responses to injury. Most neuronal culture methods employ nonphysiological conditions with regard to metabolic parameters. Standard neuronal cell culture is performed at ambient (21%) oxygen levels, whereas actual tissue oxygen levels in the mammalian brain range from 1% to 5%. In this study, we examined the consequences of oxygen level on the viability and metabolism of primary cultures of cortical neurons. Our results indicate that physiological oxygen level (5% O(2)) has a beneficial effect on cortical neuronal survival and mitochondrial function in vitro. Moreover, oxygen level affects metabolic fluxes: glucose uptake and glycolysis was enhanced at physiological oxygen level, whereas glucose oxidation and fatty acid oxidation were reduced. Adenosine monophosphate-activated protein kinase (AMPK) was more activated in 5% O(2) and appears to play a role in these metabolic effects. Inhibiting AMPK activity with compound C decreased glucose uptake, intracellular ATP level, and viability in neurons cultured in 5% O(2). These data indicate that oxygen level is an important parameter to consider when modeling neuronal responses to stress in vitro.


Subject(s)
Cerebral Cortex/physiology , Energy Metabolism/physiology , Models, Neurological , Neurons/physiology , Oxygen Consumption/physiology , Animals , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Female , Pregnancy , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
10.
J Environ Public Health ; 2022: 7492655, 2022.
Article in English | MEDLINE | ID: mdl-36089968

ABSTRACT

The current new round of scientific and technological revolution represented by artificial intelligence is rapidly driving a new wave of development of the times, and the rapid iterative update of science and technology is triggering new changes in educational concepts and educational thinking methods. Only by deeply understanding the reshaping of education concept, teaching concept, and learning concept by the new generation of scientific and technological revolution, as well as the major opportunities and challenges brought to education, can we understand the future direction of the development of ideological and political education for college students. This study takes the ideological and political education and teaching of college students as the research object. It begins by defining artificial intelligence and the ideological and political education of college students and analyzes the new concepts of precise individualization, intelligent teaching, and evaluation brought by artificial intelligence to the ideological and political education of college students. Then, it selects the students who have studied ideological and political education network resources as the empirical objects, designs a questionnaire based on the emotional characteristics of the resources, implements a questionnaire survey, uses the Stata software to conduct a correlation analysis on the acceptance of the students, and finally verifies the resources. Combined with empirical results, this article analyzes the influence of emotional characteristics of resources on students' acceptance, reveals the carrier role of ideological and political network resources in school emotional education and students through mirror theory and student response theory, respectively, and establishes the principle of graded reading guidance.


Subject(s)
Artificial Intelligence , Attitude , Educational Status , Humans , Students , Universities
11.
Mol Cell Biol ; 27(20): 7236-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17709399

ABSTRACT

The size of an organ must be tightly controlled so that it fits within an organism. The mammalian lens is a relatively simple organ composed of terminally differentiated, amitotic lens fiber cells capped on the anterior surface by a layer of immature, mitotic epithelial cells. The proliferation of lens epithelial cells fuels the growth of the lens, thus controling the size of the lens. We report that the Notch signaling pathway defines the boundary between proliferation and differentiation in the developing lens. The loss of Notch signaling results in the loss of epithelial cells to differentiation and a much smaller lens. We found that the Notch effector Herp2 is expressed in lens epithelium and directly suppresses p57Kip2 expression, providing a molecular link between Notch signaling and the cell cycle control machinery during lens development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Gene Expression Regulation , Lens, Crystalline , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , COS Cells , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Proliferation , Chlorocebus aethiops , Cyclin-Dependent Kinase Inhibitor p57/genetics , Epithelial Cells/cytology , Epithelial Cells/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein , In Situ Hybridization , Lens, Crystalline/anatomy & histology , Lens, Crystalline/physiology , Mice , Mice, Knockout , Phenotype , Receptors, Notch/genetics , Repressor Proteins/genetics
12.
Front Genet ; 11: 906, 2020.
Article in English | MEDLINE | ID: mdl-32922438

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most prevalent life-threatening human cancers and the leading cause of cancer-related mortality, with increased global incidence within the last decade. Identification of effective diagnostic and prognostic biomarkers would enable reliable risk stratification and efficient screening of high-risk patients, thereby facilitating clinical decision-making. Herein, we performed a comprehensive, robust DNA methylation analysis based on genome-wide DNA methylation profiling. We constructed a diagnostic signature with five DNA methylation markers, which precisely distinguished HCC patients from normal controls. Cox regression and LASSO analysis were applied to construct a prognostic signature with four DNA methylation markers. A one-to-one correlation analysis was carried out between genes of the whole genome and our prognostic signature. Exploration of the biological function and the role of the underlying significantly correlated genes was conducted. A mixed dataset of 463 HCC patients and 253 normal controls, derived from six independent datasets, was used to valid the diagnostic signature. Results showed a specificity of 96.84% and sensitivity of 96.77%. Class scores for the diagnostic signature were significantly different between normal controls, individuals with liver diseases, and HCC patients. The present signature has the potential to serve as a biomarker to monitor health in normal controls. Additionally, HCC patients were successfully separated into low-risk and high-risk groups by the prognostic signature, with a better prognosis for patients in the low-risk group. Kaplan-Meier and ROC analysis confirmed that the prognostic signature performed well. We found eight of the top ten genes to positively correlate with risk scores of the prognostic signature, and to be involved in cell cycle regulation. This eight-gene panel also served as a prognostic signature. The robust evidence presented in this study therefore demonstrates the effectiveness of the prognostic signature. In summary, we constructed diagnostic and prognostic signatures, which have potential for use in diagnosis, surveillance, and prognostic prediction for HCC patients. Eight genes that were significantly and positively correlated with the prognostic signature were strongly associated with cell cycle processes. Therefore, the prognostic signature can be used as a guide by which to measure responsiveness to cell-cycle-targeting agents.

13.
Genome Biol ; 19(1): 163, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30333049

ABSTRACT

Current single-cell RNA-seq approaches are hindered by preamplification bias, loss of strand of origin information, and the inability to observe small-RNA and mRNA dual transcriptomes. Here, we introduce a single-cell holo-transcriptome sequencing (Holo-Seq) that overcomes all three hurdles. Holo-Seq has the same quantitative accuracy and uniform coverage with a complete strand of origin information as bulk RNA-seq. Most importantly, Holo-Seq can simultaneously observe small RNAs and mRNAs in a single cell. Furthermore, we acquire small RNA and mRNA dual transcriptomes of 32 human hepatocellular carcinoma single cells, which display the genome-wide super-enhancer activity and hepatic neoplasm kinetics of these cells.


Subject(s)
Sequence Analysis, RNA/methods , Single-Cell Analysis , Transcriptome/genetics , Animals , HEK293 Cells , Humans , Introns/genetics , MCF-7 Cells , Male , Mice, Inbred C57BL , Middle Aged , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
PLoS One ; 13(11): e0206844, 2018.
Article in English | MEDLINE | ID: mdl-30399165

ABSTRACT

BACKGROUND: Key regulators of developmental processes can be prioritized through integrated analysis of ChIP-Seq data of master transcriptional factors (TFs) such as Nanog and Oct4, active histone modifications (HMs) such as H3K4me3 and H3K27ac, and repressive HMs such as H3K27me3. Recent studies show that broad enrichment signals such as super-enhancers and broad H3K4me3 enrichment signals play more dominant roles than short enrichment signals of the master TFs and H3K4me3 in epigenetic regulatory mechanism. Besides the broad enrichment signals, up to ten thousands of short enrichment signals of these TFs and HMs exist in genome. Prioritization of these broad enrichment signals from ChIP-Seq data is a prerequisite for such integrated analysis. RESULTS: Here, we present a method named Clustering-Local-Unique-Enriched-Signals (CLUES), which uses an adaptive-size-windows strategy to identify enriched regions (ERs) and cluster them into broad enrichment signals. Tested on 62 ENCODE ChIP-Seq datasets of Ctcf and Nrsf, CLUES performs equally well as MACS2 regarding prioritization of ERs with the TF's motif. Tested on 165 ENCODE ChIP-Seq datasets of H3K4me3, H3K27me3, and H3K36me3, CLUES performs better than existing algorithms on prioritizing broad enrichment signals implicating cell functions influenced by epigenetic regulatory mechanism in cells. Most importantly, CLUES helps to confirm several novel regulators of mouse ES cell self-renewal and pluripotency through integrated analysis of prioritized broad enrichment signals of H3K4me3, H3K27me3, Nanog and Oct4 with the support of a CRISPR/Cas9 negative selection genetic screen. CONCLUSIONS: CLUES holds promise for prioritizing broad enrichment signals from ChIP-Seq data. The download site for CLUES is https://github.com/Wuchao1984/CLUESv1.


Subject(s)
Cell Self Renewal/genetics , Embryonic Stem Cells , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Animals , CRISPR-Cas Systems/genetics , Chromatin Immunoprecipitation , Histone Code/genetics , Mice , Promoter Regions, Genetic , Protein Processing, Post-Translational , Regulatory Sequences, Nucleic Acid
16.
Nat Commun ; 8: 15021, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28443643

ABSTRACT

TGF-ß is pro-metastatic for the late-stage breast cancer cells. Despite recent progress, the regulation of TGF-ß type II receptor remains uncertain. Here we report that FAF1 destabilizes TßRII on the cell surface by recruiting the VCP/E3 ligase complex, thereby limiting excessive TGF-ß response. Importantly, activated AKT directly phosphorylates FAF1 at Ser 582, which disrupts the FAF1-VCP complex and reduces FAF1 at the plasma membrane. The latter results in an increase in TßRII at the cell surface that promotes both TGF-ß-induced SMAD and non-SMAD signalling. We uncover a metastasis suppressing role for FAF1 through analyses of FAF1-knockout animals, various in vitro and in vivo models of epithelial-to-mesenchymal transition and metastasis, an MMTV-PyMT transgenic mouse model of mammary tumour progression and clinical breast cancer samples. These findings describe a previously uncharacterized mechanism by which TßRII is tightly controlled. Together, we reveal how SMAD and AKT pathways interact to confer pro-oncogenic responses to TGF-ß.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Transforming Growth Factor beta/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Metastasis , Phosphorylation , Receptor, Transforming Growth Factor-beta Type II , Transplantation, Heterologous
17.
Nat Commun ; 8(1): 1805, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180626

ABSTRACT

Macrophages, dendritic cells and other innate immune cells are involved in inflammation and host defense against infection. Metabolic shifts in mitochondrial dynamics may be involved in Toll-like receptor agonist-mediated inflammatory responses and immune cell polarization. However, whether the mitochondrial morphology in myeloid immune cells affects anti-tumor immunity is unclear. Here we show that FAM73b, a mitochondrial outer membrane protein, has a pivotal function in Toll-like receptor-regulated mitochondrial morphology switching from fusion to fission. Switching to mitochondrial fission via ablation of Fam73b (also known as Miga2) promotes IL-12 production. In tumor-associated macrophages, this switch results in T-cell activation and enhances anti-tumor immunity. We also show that the mitochondrial morphology affects Parkin expression and its recruitment to mitochondria. Parkin controls the stability of the downstream CHIP-IRF1 axis through proteolysis. Our findings identify mechanisms associated with mitochondrial dynamics that control anti-tumor immune responses and that are potential targets for cancer immunotherapy.


Subject(s)
Immunity, Innate , Mitochondria/metabolism , Mitochondrial Dynamics/immunology , Neoplasms/immunology , Signal Transduction/immunology , Animals , Female , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interleukin-12/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/metabolism , Proteolysis , T-Lymphocytes/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
18.
Nat Commun ; 8(1): 2116, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235476

ABSTRACT

Metastasis is the main cause of death in cancer patients. TGF-ß is pro-metastatic for malignant cancer cells. Here we report a loss-of-function screen in mice with metastasis as readout and identify OTUD1 as a metastasis-repressing factor. OTUD1-silenced cancer cells show mesenchymal and stem-cell-like characteristics. Further investigation reveals that OTUD1 directly deubiquitinates the TGF-ß pathway inhibitor SMAD7 and prevents its degradation. Moreover, OTUD1 cleaves Lysine 33-linked poly-ubiquitin chains of SMAD7 Lysine 220, which exposes the SMAD7 PY motif, enabling SMURF2 binding and subsequent TßRI turnover at the cell surface. Importantly, OTUD1 is lost in multiple types of human cancers and loss of OTUD1 increases metastasis in intracardial xenograft and orthotopic transplantation models, and correlates with poor prognosis among breast cancer patients. High levels of OTUD1 inhibit cancer stemness and shut off metastasis. Thus, OTUD1 represses breast cancer metastasis by mitigating TGF-ß-induced pro-oncogenic responses via deubiquitination of SMAD7.


Subject(s)
Breast Neoplasms/metabolism , Smad7 Protein/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , Doxycycline/pharmacology , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , RNA Interference , Smad7 Protein/genetics , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays
19.
Elife ; 52016 Sep 09.
Article in English | MEDLINE | ID: mdl-27612383

ABSTRACT

Starvation induces sustained increase in locomotion, which facilitates food localization and acquisition and hence composes an important aspect of food-seeking behavior. We investigated how nutritional states modulated starvation-induced hyperactivity in adult Drosophila. The receptor of the adipokinetic hormone (AKHR), the insect analog of glucagon, was required for starvation-induced hyperactivity. AKHR was expressed in a small group of octopaminergic neurons in the brain. Silencing AKHR+ neurons and blocking octopamine signaling in these neurons eliminated starvation-induced hyperactivity, whereas activation of these neurons accelerated the onset of hyperactivity upon starvation. Neither AKHR nor AKHR+ neurons were involved in increased food consumption upon starvation, suggesting that starvation-induced hyperactivity and food consumption are independently regulated. Single cell analysis of AKHR+ neurons identified the co-expression of Drosophila insulin-like receptor (dInR), which imposed suppressive effect on starvation-induced hyperactivity. Therefore, insulin and glucagon signaling exert opposite effects on starvation-induced hyperactivity via a common neural target in Drosophila.

20.
J Bone Miner Res ; 31(10): 1888-1898, 2016 10.
Article in English | MEDLINE | ID: mdl-27128386

ABSTRACT

The canonical Wnt/ß-catenin signaling pathway plays a pivotal role and is essentially required for the osteoblast differentiation and bone formation. In this study, we found ubiquitin-specific peptidase 4 (USP4) to strongly inhibit the Wnt/ß-catenin signaling by removing Lysine-63 linked poly-ubiquitin chain from Dishevelled (Dvl). Ectopic expression of USP4 promoted ß-catenin poly-ubiquitination and thus inhibited Wnt-induced accumulation of cytosolic ß-catenin and counteracted Wnt-induced transcriptional activity. Moreover, USP4 knockdown or USP4 knockout led to an increase in the active ß-catenin levels and in activation of Wnt/ß-catenin-induced transcription. Functional studies in C2C12 myoblasts and KS483 osteoprogenitor cells showed that ectopic expression of USP4 resulted in impaired activation of endogenous Wnt3a-induced genes and decreased osteoblast differentiation and mineralization, whereas USP4 depletion showed the opposite effect. These results identify USP4 as a novel regulator of Dvl in Wnt/ß-catenin signal and show its involvement in Wnt3a-induced osteoblast differentiation. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Cell Differentiation/physiology , Dishevelled Proteins/metabolism , Osteoblasts/metabolism , Polyubiquitin/metabolism , Ubiquitin-Specific Proteases/metabolism , Wnt Signaling Pathway/physiology , Dishevelled Proteins/genetics , HeLa Cells , Humans , Polyubiquitin/genetics , Ubiquitin-Specific Proteases/genetics , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL