Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(21)2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33167521

ABSTRACT

Adipose tissue is a highly dynamic endocrine tissue and constitutes a central node in the interorgan crosstalk network through adipokines, which cause pleiotropic effects, including the modulation of angiogenesis, metabolism, and inflammation. Specifically, digestive cancers grow anatomically near adipose tissue. During their interaction with cancer cells, adipocytes are reprogrammed into cancer-associated adipocytes and secrete adipokines to affect tumor cells. Moreover, the liver is the central metabolic hub. Adipose tissue and the liver cooperatively regulate whole-body energy homeostasis via adipokines. Obesity, the excessive accumulation of adipose tissue due to hyperplasia and hypertrophy, is currently considered a global epidemic and is related to low-grade systemic inflammation characterized by altered adipokine regulation. Obesity-related digestive diseases, including gastroesophageal reflux disease, Barrett's esophagus, esophageal cancer, colon polyps and cancer, non-alcoholic fatty liver disease, viral hepatitis-related diseases, cholelithiasis, gallbladder cancer, cholangiocarcinoma, pancreatic cancer, and diabetes, might cause specific alterations in adipokine profiles. These patterns and associated bases potentially contribute to the identification of prognostic biomarkers and therapeutic approaches for the associated digestive diseases. This review highlights important findings about altered adipokine profiles relevant to digestive diseases, including hepatic, pancreatic, gastrointestinal, and biliary tract diseases, with a perspective on clinical implications and mechanistic explorations.


Subject(s)
Adipokines/metabolism , Adipokines/physiology , Digestive System Diseases/metabolism , Adipocytes/metabolism , Adiponectin/metabolism , Adipose Tissue/metabolism , Biomarkers/metabolism , Digestive System/metabolism , Digestive System/pathology , Digestive System Diseases/physiopathology , Homeostasis , Humans , Inflammation/metabolism , Leptin/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism
2.
Nat Metab ; 2(5): 397-412, 2020 05.
Article in English | MEDLINE | ID: mdl-32440655

ABSTRACT

Human thermogenic adipose tissue mitigates metabolic disease, raising much interest in understanding its development and function. Here, we show that human thermogenic adipocytes specifically express a primate-specific long non-coding RNA, LINC00473 which is highly correlated with UCP1 expression and decreased in obesity and type-2 diabetes. LINC00473 is detected in progenitor cells, and increases upon differentiation and in response to cAMP. In contrast to other known adipocyte LincRNAs, LINC00473 shuttles out of the nucleus, colocalizes and can be crosslinked to mitochondrial and lipid droplet proteins. Up- or down- regulation of LINC00473 results in reciprocal alterations in lipolysis, respiration and transcription of genes associated with mitochondrial oxidative metabolism. Depletion of PLIN1 results in impaired cAMP-responsive LINC00473 expression and lipolysis, indicating bidirectional interactions between PLIN1, LINC00473 and mitochondrial oxidative functions. Thus, we suggest that LINC00473 is a key regulator of human thermogenic adipocyte function, and reveals a role for a LincRNA in inter-organelle communication and human energy metabolism.


Subject(s)
Adipocytes/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Thermogenesis/genetics , Thermogenesis/physiology , Adult , Aged , Aged, 80 and over , Cell Communication/genetics , Cell Communication/physiology , Cell Nucleus/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/genetics , Energy Metabolism/physiology , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression Regulation , Humans , Lipid Droplets , Male , Middle Aged , Obesity/genetics , Obesity/metabolism , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Perilipin-1/deficiency , Perilipin-1/genetics , Uncoupling Protein 1/biosynthesis , Uncoupling Protein 1/genetics , Young Adult
3.
Elife ; 82019 11 19.
Article in English | MEDLINE | ID: mdl-31741433

ABSTRACT

EGFR-mutant NSCLCs frequently respond to EGFR tyrosine kinase inhibitors (TKIs). However, the responses are not durable, and the magnitude of tumor regression is variable, suggesting the existence of genetic modifiers of EGFR dependency. Here, we applied a genome-wide CRISPR-Cas9 screening to identify genetic determinants of EGFR TKI sensitivity and uncovered putative candidates. We show that knockout of RIC8A, essential for G-alpha protein activation, enhanced EGFR TKI-induced cell death. Mechanistically, we demonstrate that RIC8A is a positive regulator of YAP signaling, activation of which rescued the EGFR TKI sensitizing phenotype resulting from RIC8A knockout. We also show that knockout of ARIH2, or other components in the Cullin-5 E3 complex, conferred resistance to EGFR inhibition, in part by promoting nascent protein synthesis through METAP2. Together, these data uncover a spectrum of previously unidentified regulators of EGFR TKI sensitivity in EGFR-mutant human NSCLC, providing insights into the heterogeneity of EGFR TKI treatment responses.


Subject(s)
CRISPR-Cas Systems , Carcinoma, Non-Small-Cell Lung/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , A549 Cells , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cullin Proteins , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Methionyl Aminopeptidases/metabolism , Mice , Mice, Nude , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcriptome , Ubiquitin-Protein Ligases/genetics , YAP-Signaling Proteins , rhoA GTP-Binding Protein/metabolism
4.
Nat Commun ; 10(1): 4184, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519875

ABSTRACT

Axin is a key scaffolding protein responsible for the formation of the ß-catenin destruction complex. Stability of Axin protein is regulated by the ubiquitin-proteasome system, and modulation of cellular concentration of Axin protein has a profound effect on Wnt/ß-catenin signaling. Although E3s promoting Axin ubiquitination have been identified, the deubiquitinase responsible for Axin deubiquitination and stabilization remains unknown. Here, we identify USP7 as a potent negative regulator of Wnt/ß-catenin signaling through CRISPR screens. Genetic ablation or pharmacological inhibition of USP7 robustly increases Wnt/ß-catenin signaling in multiple cellular systems. USP7 directly interacts with Axin through its TRAF domain, and promotes deubiquitination and stabilization of Axin. Inhibition of USP7 regulates osteoblast differentiation and adipocyte differentiation through increasing Wnt/ß-catenin signaling. Our study reveals a critical mechanism that prevents excessive degradation of Axin and identifies USP7 as a target for sensitizing cells to Wnt/ß-catenin signaling.


Subject(s)
Axin Protein/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , beta Catenin/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Axin Protein/genetics , Cell Line , Cell Line, Tumor , Flow Cytometry , HCT116 Cells , Humans , Immunoprecipitation , Mice , Osteoblasts/metabolism , Protein Stability , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitination/genetics , Ubiquitination/physiology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics
5.
Proc Natl Acad Sci U S A ; 116(36): 17970-17979, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31420514

ABSTRACT

Single-cell sequencing technologies have revealed an unexpectedly broad repertoire of cells required to mediate complex functions in multicellular organisms. Despite the multiple roles of adipose tissue in maintaining systemic metabolic homeostasis, adipocytes are thought to be largely homogenous with only 2 major subtypes recognized in humans so far. Here we report the existence and characteristics of 4 distinct human adipocyte subtypes, and of their respective mesenchymal progenitors. The phenotypes of these distinct adipocyte subtypes are differentially associated with key adipose tissue functions, including thermogenesis, lipid storage, and adipokine secretion. The transcriptomic signature of "brite/beige" thermogenic adipocytes reveals mechanisms for iron accumulation and protection from oxidative stress, necessary for mitochondrial biogenesis and respiration upon activation. Importantly, this signature is enriched in human supraclavicular adipose tissue, confirming that these cells comprise thermogenic depots in vivo, and explain previous findings of a rate-limiting role of iron in adipose tissue browning. The mesenchymal progenitors that give rise to beige/brite adipocytes express a unique set of cytokines and transcriptional regulators involved in immune cell modulation of adipose tissue browning. Unexpectedly, we also find adipocyte subtypes specialized for high-level expression of the adipokines adiponectin or leptin, associated with distinct transcription factors previously implicated in adipocyte differentiation. The finding of a broad adipocyte repertoire derived from a distinct set of mesenchymal progenitors, and of the transcriptional regulators that can control their development, provides a framework for understanding human adipose tissue function and role in metabolic disease.


Subject(s)
Adipocytes, Beige/metabolism , Adiponectin/biosynthesis , Leptin/blood , Mesenchymal Stem Cells/metabolism , Thermogenesis , Transcriptome , Adipocytes, Beige/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Female , Gene Expression Profiling , Humans , Male , Mesenchymal Stem Cells/cytology
6.
Cell Stem Cell ; 25(1): 39-53.e10, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31080135

ABSTRACT

Biliary epithelial cells (BECs) form bile ducts in the liver and are facultative liver stem cells that establish a ductular reaction (DR) to support liver regeneration following injury. Liver damage induces periportal LGR5+ putative liver stem cells that can form BEC-like organoids, suggesting that RSPO-LGR4/5-mediated WNT/ß-catenin activity is important for a DR. We addressed the roles of this and other signaling pathways in a DR by performing a focused CRISPR-based loss-of-function screen in BEC-like organoids, followed by in vivo validation and single-cell RNA sequencing. We found that BECs lack and do not require LGR4/5-mediated WNT/ß-catenin signaling during a DR, whereas YAP and mTORC1 signaling are required for this process. Upregulation of AXIN2 and LGR5 is required in hepatocytes to enable their regenerative capacity in response to injury. Together, these data highlight heterogeneity within the BEC pool, delineate signaling pathways involved in a DR, and clarify the identity and roles of injury-induced periportal LGR5+ cells.


Subject(s)
Acute Lung Injury/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Bile Ducts/pathology , Cell Cycle Proteins/metabolism , Epithelial Cells/physiology , Induced Pluripotent Stem Cells/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Axin Protein/genetics , Axin Protein/metabolism , Cell Cycle Proteins/genetics , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Models, Animal , Humans , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Pyridines/toxicity , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Thrombospondins/genetics , Thrombospondins/metabolism , Wnt Signaling Pathway , YAP-Signaling Proteins
7.
Cell Rep ; 27(2): 616-630.e6, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970262

ABSTRACT

Human pluripotent stem cells (hPSCs) generate a variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited by technical challenges. We developed a scalable and renewable Cas9 and sgRNA-hPSC library in which loss-of-function mutations can be induced at will. Our inducible mutant hPSC library can be used for multiple genome-wide CRISPR screens in a variety of hPSC-induced cell types. As proof of concept, we performed three screens for regulators of properties fundamental to hPSCs: their ability to self-renew and/or survive (fitness), their inability to survive as single-cell clones, and their capacity to differentiate. We identified the majority of known genes and pathways involved in these processes, as well as a plethora of genes with unidentified roles. This resource will increase the understanding of human development and genetics. This approach will be a powerful tool to identify disease-modifying genes and pathways.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Testing/methods , Genome/genetics , Pluripotent Stem Cells/metabolism , Humans
8.
J Biol Chem ; 294(10): 3359-3366, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30647128

ABSTRACT

Bile acids are critical metabolites in the gastrointestinal tract and contribute to maintaining intestinal immune homeostasis through cross-talk with the gut microbiota. The conversion of bile acids by the gut microbiome is now recognized as a factor affecting both host metabolism and immune responses, but its physiological roles remain unclear. We conducted a screen for microbiome metabolites that would function as inflammasome activators and herein report the identification of 12-oxo-lithocholic acid (BAA485), a potential microbiome-derived bile acid metabolite. We demonstrate that the more potent analogue 11-oxo-12S-hydroxylithocholic acid methyl ester (BAA473) can induce secretion of interleukin-18 (IL-18) through activation of the inflammasome in both myeloid and intestinal epithelial cells. Using a genome-wide CRISPR screen with compound induced pyroptosis in THP-1 cells, we identified that inflammasome activation by BAA473 is pyrin-dependent (MEFV). To our knowledge, the bile acid analogues BAA485 and BAA473 are the first small molecule activators of the pyrin inflammasome. We surmise that pyrin inflammasome activation through microbiota-modified bile acid metabolites such as BAA473 and BAA485 plays a role in gut microbiota regulated intestinal immune response. The discovery of these two bioactive compounds may help to further unveil the importance of pyrin in gut homeostasis and autoimmune diseases.


Subject(s)
Bile Acids and Salts/immunology , Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Inflammasomes/immunology , Intestinal Mucosa/immunology , Pyrin/immunology , Bile Acids and Salts/chemistry , Humans , Myeloid Cells/immunology , THP-1 Cells
9.
Proc Natl Acad Sci U S A ; 115(44): E10362-E10369, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30297426

ABSTRACT

Wnt/ß-catenin signaling plays pivotal roles in cell proliferation and tissue homeostasis by maintaining somatic stem cell functions. The mammalian target of rapamycin (mTOR) signaling functions as an integrative rheostat that orchestrates various cellular and metabolic activities that shape tissue homeostasis. Whether these two fundamental signaling pathways couple to exert physiological functions still remains mysterious. Using a genome-wide CRISPR-Cas9 screening, we discover that mTOR complex 1 (mTORC1) signaling suppresses canonical Wnt/ß-catenin signaling. Deficiency in tuberous sclerosis complex 1/2 (TSC1/2), core negative regulators of mTORC1 activity, represses Wnt/ß-catenin target gene expression, which can be rescued by RAD001. Mechanistically, mTORC1 signaling regulates the cell surface level of Wnt receptor Frizzled (FZD) in a Dishevelled (DVL)-dependent manner by influencing the association of DVL and clathrin AP-2 adaptor. Sustained mTORC1 activation impairs Wnt/ß-catenin signaling and causes loss of stemness in intestinal organoids ex vivo and primitive intestinal progenitors in vivo. Wnt/ß-catenin-dependent liver metabolic zonation gene expression program is also down-regulated by mTORC1 activation. Our study provides a paradigm that mTORC1 signaling cell autonomously regulates Wnt/ß-catenin pathway to influence stem cell maintenance.


Subject(s)
Frizzled Receptors/metabolism , Receptors, Wnt/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Cell Line , Dishevelled Proteins/metabolism , Down-Regulation/physiology , Gene Expression/physiology , HEK293 Cells , Humans , Mice
10.
Mol Cell ; 72(1): 37-47.e4, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30197296

ABSTRACT

Adenomatous polyposis coli (APC) and Axin are core components of the ß-catenin destruction complex. How APC's function is regulated and whether Wnt signaling influences the direct APC-Axin interaction to inhibit the ß-catenin destruction complex is not clear. Through a CRISPR screen of ß-catenin stability, we have identified ICAT, a polypeptide previously known to block ß-catenin-TCF interaction, as a natural inhibitor of APC. ICAT blocks ß-catenin-APC interaction and prevents ß-catenin-mediated APC-Axin interaction, enhancing stabilization of ß-catenin in cells harboring truncated APC or stimulated with Wnt, but not in cells deprived of a Wnt signal. Using ICAT as a tool to disengage ß-catenin-mediated APC-Axin interaction, we demonstrate that Wnt quickly inhibits the direct interaction between APC and Axin. Our study highlights an important scaffolding function of ß-catenin in the assembly of the destruction complex and suggests Wnt-inhibited APC-Axin interaction as a mechanism of Wnt-dependent inhibition of the destruction complex.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Interaction Domains and Motifs/genetics , beta Catenin/genetics , Adaptor Proteins, Signal Transducing , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Axin Protein/genetics , Humans , Protein Stability , Transcription Factor 7-Like 1 Protein/genetics , Wnt Signaling Pathway/genetics
11.
EMBO Rep ; 19(9)2018 09.
Article in English | MEDLINE | ID: mdl-30126924

ABSTRACT

Autophagy maintains cellular homeostasis by targeting damaged organelles, pathogens, or misfolded protein aggregates for lysosomal degradation. The autophagic process is initiated by the formation of autophagosomes, which can selectively enclose cargo via autophagy cargo receptors. A machinery of well-characterized autophagy-related proteins orchestrates the biogenesis of autophagosomes; however, the origin of the required membranes is incompletely understood. Here, we have applied sensitized pooled CRISPR screens and identify the uncharacterized transmembrane protein TMEM41B as a novel regulator of autophagy. In the absence of TMEM41B, autophagosome biogenesis is stalled, LC3 accumulates at WIPI2- and DFCP1-positive isolation membranes, and lysosomal flux of autophagy cargo receptors and intracellular bacteria is impaired. In addition to defective autophagy, TMEM41B knockout cells display significantly enlarged lipid droplets and reduced mobilization and ß-oxidation of fatty acids. Immunostaining and interaction proteomics data suggest that TMEM41B localizes to the endoplasmic reticulum (ER). Taken together, we propose that TMEM41B is a novel ER-localized regulator of autophagosome biogenesis and lipid mobilization.


Subject(s)
Autophagy/physiology , Lipid Mobilization/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Autophagosomes/metabolism , Autophagy/genetics , Autophagy-Related Proteins/metabolism , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Endoplasmic Reticulum/metabolism , Fatty Acids/metabolism , Gene Knockout Techniques , HeLa Cells , Homeostasis , Humans , Lentivirus , Lipid Droplets/metabolism , Lipid Mobilization/genetics , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism
12.
Nat Med ; 24(7): 939-946, 2018 07.
Article in English | MEDLINE | ID: mdl-29892062

ABSTRACT

CRISPR/Cas9 has revolutionized our ability to engineer genomes and conduct genome-wide screens in human cells1-3. Whereas some cell types are amenable to genome engineering, genomes of human pluripotent stem cells (hPSCs) have been difficult to engineer, with reduced efficiencies relative to tumour cell lines or mouse embryonic stem cells3-13. Here, using hPSC lines with stable integration of Cas9 or transient delivery of Cas9-ribonucleoproteins (RNPs), we achieved an average insertion or deletion (indel) efficiency greater than 80%. This high efficiency of indel generation revealed that double-strand breaks (DSBs) induced by Cas9 are toxic and kill most hPSCs. In previous studies, the toxicity of Cas9 in hPSCs was less apparent because of low transfection efficiency and subsequently low DSB induction3. The toxic response to DSBs was P53/TP53-dependent, such that the efficiency of precise genome engineering in hPSCs with a wild-type P53 gene was severely reduced. Our results indicate that Cas9 toxicity creates an obstacle to the high-throughput use of CRISPR/Cas9 for genome engineering and screening in hPSCs. Moreover, as hPSCs can acquire P53 mutations14, cell replacement therapies using CRISPR/Cas9-enginereed hPSCs should proceed with caution, and such engineered hPSCs should be monitored for P53 function.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Genetic Engineering , Pluripotent Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Breaks, Double-Stranded , Gene Deletion , Humans , RNA, Guide, Kinetoplastida/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , fas Receptor/genetics , fas Receptor/metabolism
13.
J Cell Biol ; 217(6): 1941-1955, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29588376

ABSTRACT

The multiciliated cell (MCC) is an evolutionarily conserved cell type, which in vertebrates functions to promote directional fluid flow across epithelial tissues. In the conducting airway, MCCs are generated by basal stem/progenitor cells and act in concert with secretory cells to perform mucociliary clearance to expel pathogens from the lung. Studies in multiple systems, including Xenopus laevis epidermis, murine trachea, and zebrafish kidney, have uncovered a transcriptional network that regulates multiple steps of multiciliogenesis, ultimately leading to an MCC with hundreds of motile cilia extended from their apical surface, which beat in a coordinated fashion. Here, we used a pool-based short hairpin RNA screening approach and identified TRRAP, an essential component of multiple histone acetyltransferase complexes, as a central regulator of MCC formation. Using a combination of immunofluorescence, signaling pathway modulation, and genomic approaches, we show that (a) TRRAP acts downstream of the Notch2-mediated basal progenitor cell fate decision and upstream of Multicilin to control MCC differentiation; and (b) TRRAP binds to the promoters and regulates the expression of a network of genes involved in MCC differentiation and function, including several genes associated with human ciliopathies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cilia/metabolism , Nuclear Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Lineage , Epigenesis, Genetic , Epithelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Lung/cytology , RNA, Small Interfering/metabolism , Receptor, Notch2 , Signal Transduction , Transcription Factors
14.
Sci Rep ; 7: 42728, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205648

ABSTRACT

Chemogenomic profiling is a powerful and unbiased approach to elucidate pharmacological targets and the mechanism of bioactive compounds. Until recently, genome-wide, high-resolution experiments of this nature have been limited to fungal systems due to lack of mammalian genome-wide deletion collections. With the example of a novel nicotinamide phosphoribosyltransferase (NAMPT) inhibitor, we demonstrate that the CRISPR/Cas9 system enables the generation of transient homo- and heterozygous deletion libraries and allows for the identification of efficacy targets and pathways mediating hypersensitivity and resistance relevant to the compound mechanism of action.


Subject(s)
CRISPR-Cas Systems , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Cells, Cultured , Enzyme Inhibitors/chemistry , Gene Deletion , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Pharmacogenomic Testing/methods
15.
J Biol Chem ; 291(29): 15256-66, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27231341

ABSTRACT

YAP signaling pathway plays critical roles in tissue homeostasis, and aberrant activation of YAP signaling has been implicated in cancers. To identify tractable targets of YAP pathway, we have performed a pathway-based pooled CRISPR screen and identified tankyrase and its associated E3 ligase RNF146 as positive regulators of YAP signaling. Genetic ablation or pharmacological inhibition of tankyrase prominently suppresses YAP activity and YAP target gene expression. Using a proteomic approach, we have identified angiomotin family proteins, which are known negative regulators of YAP signaling, as novel tankyrase substrates. Inhibition of tankyrase or depletion of RNF146 stabilizes angiomotins. Angiomotins physically interact with tankyrase through a highly conserved motif at their N terminus, and mutation of this motif leads to their stabilization. Tankyrase inhibitor-induced stabilization of angiomotins reduces YAP nuclear translocation and decreases downstream YAP signaling. We have further shown that knock-out of YAP sensitizes non-small cell lung cancer to EGFR inhibitor Erlotinib. Tankyrase inhibitor, but not porcupine inhibitor, which blocks Wnt secretion, enhances growth inhibitory activity of Erlotinib. This activity is mediated by YAP inhibition and not Wnt/ß-catenin inhibition. Our data suggest that tankyrase inhibition could serve as a novel strategy to suppress YAP signaling for combinatorial targeted therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Phosphoproteins/antagonists & inhibitors , Tankyrases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Angiomotins , Antineoplastic Agents/pharmacology , CRISPR-Cas Systems , Cell Line, Tumor , Down-Regulation , Erlotinib Hydrochloride/pharmacology , Gene Knockout Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microfilament Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Interaction Domains and Motifs , Protein Stability/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tankyrases/chemistry , Tankyrases/genetics , Transcription Factors , Ubiquitin-Protein Ligases/metabolism , YAP-Signaling Proteins
16.
J Clin Invest ; 117(10): 3007-19, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17885688

ABSTRACT

The discovery of fetal mRNA transcripts in the maternal circulation holds great promise for noninvasive prenatal diagnosis. To identify potential fetal biomarkers, we studied whole blood and plasma gene transcripts that were common to 9 term pregnant women and their newborns but absent or reduced in the mothers postpartum. RNA was isolated from peripheral or umbilical blood and hybridized to gene expression arrays. Gene expression, paired Student's t test, and pathway analyses were performed. In whole blood, 157 gene transcripts met statistical significance. These fetal biomarkers included 27 developmental genes, 5 sensory perception genes, and 22 genes involved in neonatal physiology. Transcripts were predominantly expressed or restricted to the fetus, the embryo, or the neonate. Real-time RT-PCR amplification confirmed the presence of specific gene transcripts; SNP analysis demonstrated the presence of 3 fetal transcripts in maternal antepartum blood. Comparison of whole blood and plasma samples from the same pregnant woman suggested that placental genes are more easily detected in plasma. We conclude that fetal and placental mRNA circulates in the blood of pregnant women. Transcriptional analysis of maternal whole blood identifies a unique set of biologically diverse fetal genes and has a multitude of clinical applications.


Subject(s)
Fetal Blood/chemistry , Fetus/metabolism , Gene Expression , Pregnancy/genetics , RNA, Messenger/blood , Adult , Biomarkers/blood , Female , Humans , Infant, Newborn , Polymorphism, Single Nucleotide , Pregnant Women , Reverse Transcriptase Polymerase Chain Reaction
17.
Am J Chin Med ; 30(4): 637-43, 2002.
Article in English | MEDLINE | ID: mdl-12568291

ABSTRACT

To determine the extent to which Taiwanese patients use alternative medicine, we interviewed 500 consecutive patients with chronic liver and gastrointestinal disorders at an outpatient-service. Forty-two patients were excluded due to incomplete data. The percentages of patients with chronic liver (102/269, 37.9%) and gastrointestinal (74/189, 39.2%) diseases using alternative medicine were not significantly different (p = 0.70). The patients who used alternative medicine were not statistically different in gender (p = 0.37), age (p = 0.59), education level (p = 0.83), family income (p = 0.90), or occupation (p = 0.72). Only 36% (64/176) of patients informed their doctors of their use of alternative medicine. The kinds of alternative medicine used by the 176 patients included: Chinese/herbal medicine, 169 (96%); acupuncture, 31 (18%); nutritional supplements, 22 (13%); chiropractic, 17 (10%); scratching, 14 (8%); Qigong, 13 (7%); cupping, 13 (7%); and incense ash, 3 (2%). Sixty-six percent (111/169) of patients used Chinese/herbal medicine in addition to Western allopathic medicine. Only 11% (19/169) of them believed that Chinese/herbal medicine had side effects. Our study indicates the use of alternative medicine occurs across all demographic groups in one-third of patients with chronic liver and gastrointestinal diseases at a major general hospital in Taipei. We suggest that the doctors question all patients for history of alternative therapy use.


Subject(s)
Complementary Therapies/statistics & numerical data , Gastrointestinal Diseases/therapy , Hepatitis, Chronic/therapy , Adult , Age Factors , Chronic Disease , Complementary Therapies/economics , Data Collection , Education , Female , Herbal Medicine/economics , Humans , Income , Male , Middle Aged , Occupations , Prospective Studies , Sex Factors , Socioeconomic Factors , Taiwan/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...