Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Clin Invest ; 133(20)2023 10 16.
Article in English | MEDLINE | ID: mdl-37843276

ABSTRACT

The loss of contact inhibition is a key step during carcinogenesis. The Hippo-Yes-associated protein (Hippo/YAP) pathway is an important regulator of cell growth in a cell density-dependent manner. However, how Hippo signaling senses cell density in this context remains elusive. Here, we report that high cell density induced the phosphorylation of spectrin α chain, nonerythrocytic 1 (SPTAN1), a plasma membrane-stabilizing protein, to recruit NUMB endocytic adaptor protein isoforms 1 and 2 (NUMB1/2), which further sequestered microtubule affinity-regulating kinases (MARKs) in the plasma membrane and rendered them inaccessible for phosphorylation and inhibition of the Hippo kinases sterile 20-like kinases MST1 and MST2 (MST1/2). WW45 interaction with MST1/2 was thereby enhanced, resulting in the activation of Hippo signaling to block YAP activity for cell contact inhibition. Importantly, low cell density led to SPTAN1 dephosphorylation and NUMB cytoplasmic location, along with MST1/2 inhibition and, consequently, YAP activation. Moreover, double KO of NUMB and WW45 in the liver led to appreciable organ enlargement and rapid tumorigenesis. Interestingly, NUMB isoforms 3 and 4, which have a truncated phosphotyrosine-binding (PTB) domain and are thus unable to interact with phosphorylated SPTAN1 and activate MST1/2, were selectively upregulated in liver cancer, which correlated with YAP activation. We have thus revealed a SPTAN1/NUMB1/2 axis that acts as a cell density sensor to restrain cell growth and oncogenesis by coupling external cell-cell contact signals to intracellular Hippo signaling.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Spectrin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins , Transcription Factors/metabolism , Carcinogenesis/genetics
2.
Nat Commun ; 13(1): 5565, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36138017

ABSTRACT

Microtubules play a crucial role during the establishment and maintenance of cell polarity. In fission yeast cells, the microtubule plus-end tracking proteins (+TIPs) (including the CLIP-170 homologue Tip1) regulate microtubule dynamics and also transport polarity factors to the cell cortex. Here, we show that the E3 ubiquitin ligase Dma1 plays an unexpected role in controlling polarized growth through ubiquitinating Tip1. Dma1 colocalizes with Tip1 to cortical sites at cell ends, and is required for ubiquitination of Tip1. Although the absence of dma1+ does not cause apparent polar growth defects in vegetatively growing cells, Dma1-mediated Tip1 ubiquitination is required to restrain polar growth upon DNA replication stress. This mechanism is distinct from the previously recognized calcineurin-dependent inhibition of polarized growth. In this work, we establish a link between Dma1-mediated Tip1 ubiquitination and DNA replication or DNA damage checkpoint-dependent inhibition of polarized growth in fission yeast.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Calcineurin/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Replication , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neoplasm Proteins , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
Cell ; 184(22): 5559-5576.e19, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34678143

ABSTRACT

Glucose consumption is generally increased in tumor cells to support tumor growth. Interestingly, we report that glycogen accumulation is a key initiating oncogenic event during liver malignant transformation. We found that glucose-6-phosphatase (G6PC) catalyzing the last step of glycogenolysis is frequently downregulated to augment glucose storage in pre-malignant cells. Accumulated glycogen undergoes liquid-liquid phase separation, which results in the assembly of the Laforin-Mst1/2 complex and consequently sequesters Hippo kinases Mst1/2 in glycogen liquid droplets to relieve their inhibition on Yap. Moreover, G6PC or another glycogenolysis enzyme-liver glycogen phosphorylase (PYGL) deficiency in both human and mice results in glycogen storage disease along with liver enlargement and tumorigenesis in a Yap-dependent manner. Consistently, elimination of glycogen accumulation abrogates liver growth and cancer incidence, whereas increasing glycogen storage accelerates tumorigenesis. Thus, we concluded that cancer-initiating cells adapt a glycogen storing mode, which blocks Hippo signaling through glycogen phase separation to augment tumor incidence.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Glycogen/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line , Disease Models, Animal , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Glucose-6-Phosphatase/metabolism , Glycogen Phosphorylase/metabolism , Hepatocyte Growth Factor/metabolism , Hippo Signaling Pathway , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging , Phase Transition , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proto-Oncogene Proteins/metabolism , Serine-Threonine Kinase 3/metabolism , YAP-Signaling Proteins/metabolism
4.
Nat Commun ; 12(1): 3519, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112781

ABSTRACT

TLR4 signaling plays key roles in the innate immune response to microbial infection. Innate immune cells encounter different mechanical cues in both health and disease to adapt their behaviors. However, the impact of mechanical sensing signals on TLR4 signal-mediated innate immune response remains unclear. Here we show that TLR4 signalling augments macrophage bactericidal activity through the mechanical sensor Piezo1. Bacterial infection or LPS stimulation triggers assembly of the complex of Piezo1 and TLR4 to remodel F-actin organization and augment phagocytosis, mitochondrion-phagosomal ROS production and bacterial clearance and genetic deficiency of Piezo1 results in abrogation of these responses. Mechanistically, LPS stimulates TLR4 to induce Piezo1-mediated calcium influx and consequently activates CaMKII-Mst1/2-Rac axis for pathogen ingestion and killing. Inhibition of CaMKII or knockout of either Mst1/2 or Rac1 results in reduced macrophage bactericidal activity, phenocopying the Piezo1 deficiency. Thus, we conclude that TLR4 drives the innate immune response via Piezo1 providing critical insight for understanding macrophage mechanophysiology and the host response.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Ion Channels/metabolism , Macrophages/immunology , Phagosomes/metabolism , Toll-Like Receptor 4/metabolism , Actins/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Escherichia coli Infections/immunology , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Ion Channels/genetics , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , Neuropeptides/metabolism , Phagocytosis/immunology , Phagosomes/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Serine-Threonine Kinase 3 , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
5.
Nat Commun ; 11(1): 4765, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32958780

ABSTRACT

Fatty acids (FAs) are essential nutrients, but how they are transported into cells remains unclear. Here, we show that FAs trigger caveolae-dependent CD36 internalization, which in turn delivers FAs into adipocytes. During the process, binding of FAs to CD36 activates its downstream kinase LYN, which phosphorylates DHHC5, the palmitoyl acyltransferase of CD36, at Tyr91 and inactivates it. CD36 then gets depalmitoylated by APT1 and recruits another tyrosine kinase SYK to phosphorylate JNK and VAVs to initiate endocytic uptake of FAs. Blocking CD36 internalization by inhibiting APT1, LYN or SYK abolishes CD36-dependent FA uptake. Restricting CD36 at either palmitoylated or depalmitoylated state eliminates its FA uptake activity, indicating an essential role of dynamic palmitoylation of CD36. Furthermore, blocking endocytosis by targeting LYN or SYK inhibits CD36-dependent lipid droplet growth in adipocytes and high-fat-diet induced weight gain in mice. Our study has uncovered a dynamic palmitoylation-regulated endocytic pathway to take up FAs.


Subject(s)
CD36 Antigens/metabolism , Endocytosis/physiology , Fatty Acids/metabolism , Lipoylation , 3T3-L1 Cells , Acyltransferases/metabolism , Adipocytes/metabolism , Animals , CD36 Antigens/deficiency , CD36 Antigens/genetics , Caveolae/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Humans , Lipid Droplets/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Obesity/drug therapy , Phosphorylation , Signal Transduction , Syk Kinase/antagonists & inhibitors , Syk Kinase/metabolism , Weight Gain/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
6.
Nanomedicine ; 21: 102047, 2019 10.
Article in English | MEDLINE | ID: mdl-31271877

ABSTRACT

Drug delivery nanocarriers based on magnetic nanoparticles have attracted increasing attention due to their potential applications in magnetic resonance imaging, photodynamic therapy and targeted drug delivery. Herein, we have fabricated the multifunctional co-loaded magnetic nanocapsules (MNCPs) using a microemulsion process for enhancing targeted magnetic resonance imaging and in vivo photodynamic therapy. MNCPs were synthesized by co-loading Co@Mn magnetic nanoparticles and chlorin e6 into the matrix of an amphiphilic polymer, and further surface covalently coupled with target molecules. This work demonstrates that MNCPs have uniform sizes (dc: ~150 nm), favorable biocompatibility, long-term stability, excellent T2 relaxation values, and high drug loading efficiency. These advantages offer MNCPs successfully applied in targeted magnetic resonance imaging, real-time fluorescent labeling, and photodynamic therapy. The research results will contribute to rationally design novel nano-platform and provide a promising approach for further clinical integration of diagnosis and treatment in the near future.


Subject(s)
Biocompatible Materials/pharmacology , Drug Delivery Systems , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Biocompatible Materials/chemistry , Chlorophyllides , Cobalt/chemistry , Cobalt/pharmacology , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Humans , Manganese/chemistry , Manganese/pharmacology , Photochemotherapy/methods , Porphyrins/chemistry , Porphyrins/pharmacology
7.
Nat Commun ; 10(1): 755, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765703

ABSTRACT

Reactive oxygen species (ROS) production in phagocytes is a major defense mechanism against pathogens. However, the cellular self-protective mechanism against such potential damage from oxidative stress remains unclear. Here we show that the kinases Mst1 and Mst2 (Mst1/2) sense ROS and maintain cellular redox balance by modulating the stability of antioxidant transcription factor Nrf2. Site-specific ROS release recruits Mst1/2 from the cytosol to the phagosomal or mitochondrial membrane, with ROS subsequently activating Mst1/2 to phosphorylate kelch like ECH associated protein 1 (Keap1) and prevent Keap1 polymerization, thereby blocking Nrf2 ubiquitination and degradation to protect cells against oxidative damage. Treatment with the antioxidant N-acetylcysteine disrupts ROS-induced interaction of Mst1/2 with phagosomes or mitochondria, and thereby diminishes the Mst-Nrf2 signal. Consistently, loss of Mst1/2 results in increased oxidative injury, phagocyte ageing and death. Thus, our results identify the Mst-Nrf2 axis as an important ROS-sensing and antioxidant mechanism during an antimicrobial response.


Subject(s)
Macrophages/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Cellular Senescence , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Protein Serine-Threonine Kinases/genetics , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Serine-Threonine Kinase 3 , Signal Transduction/genetics , THP-1 Cells
8.
Dev Cell ; 48(4): 460-474.e9, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30745141

ABSTRACT

The external factors that modulate Hippo signaling remain elusive. Here, we report that FGF15 activates Hippo signaling to suppress bile acid metabolism, liver overgrowth, and tumorigenesis. FGF15 is induced by FXR in ileal enterocytes in response to increased amounts of intestinal bile. We found that circulating enterohepatic FGF15 stimulates hepatic receptor FGFR4 to recruit and phosphorylate NF2, which relieves the inhibitory effect of Raf on the Hippo kinases Mst1/2, thereby switching FGFR4's role from pro-oncogenic to anti-tumor signaling. The activated Mst1/2 subsequently phosphorylates and stabilizes SHP to downregulate the key bile acid-synthesis enzyme Cyp7a1 expression, thereby limiting bile acid synthesis. In contrast, Mst1/2 deficiency impairs bile acid metabolism and remarkably increases Cyp7a1 expression and bile acid production. Importantly, pharmacological depletion of intestinal bile abrogates Mst1/2-mutant-driven liver overgrowth and oncogenesis. Therefore, FGF15-Hippo signaling along the gut-liver axis acts as a sensor of bile acid availability to restrain liver size and tumorigenesis.


Subject(s)
Bile Acids and Salts/metabolism , Carcinogenesis/metabolism , Fibroblast Growth Factors/metabolism , Liver Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Hippo Signaling Pathway , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Mice, Transgenic , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Signal Transduction/physiology
9.
Methods Mol Biol ; 1893: 353-370, 2019.
Article in English | MEDLINE | ID: mdl-30565146

ABSTRACT

The major role of Hippo signaling is to inhibit their downstream effectors YAP/TAZ for organ size control during development and regeneration (Nat Rev Drug Discov 13(1):63-79, 2014; Dev Cell 19(4):491-505, 2010; Cell 163(4):811-828, 2015). We and others have demonstrated that the genetic disruption of kinases Mst1 and Mst2 (Mst1/2), the core components of Hippo signaling, results in YAP activation and sustained liver growth, thereby leading to an eight- to tenfold increase in liver size within 3 months and occurrence of liver cancer within 5 months (Curr Biol 17(23):2054-2060, 2007; Cancer Cell 16(5):425-438, 2009; Cell 130(6):1120-1133, 2007; Cancer Cell 31(5):669-684 e667, 2017; Nat Commun 6:6239, 2015; Cell Rep 3(5):1663-1677, 2013). XMU-MP-1, an Mst1/2 inhibitor, is able to augment mouse liver and intestinal repair and regeneration in both acute and chronic injury mouse models (Sci Transl Med 8:352ra108, 2016).In addition, YAP-deficient mice show an impaired intestinal regenerative response after DSS treatment or gamma irradiation (Proc Natl Acad Sci U S A 108(49):E1312-1320, 2011; Nature 493(7430):106-110, 2013; Genes Dev 24(21):2383-2388, 2010; J Vis Exp (111), 2010). IBS008738, a TAZ activator, facilitates muscle repair after cardiotoxin-induced muscle injury (Mol Cell Biol. 2014;34(9):1607-21). Deletion of Salvador (Sav) in mouse hearts enhances cardiomyocyte regeneration with reduced fibrosis and recovery of pumping function after myocardial infarction (MI) or resection of mouse cardiac apex (Development 140(23):4683-4690, 2013; Sci Signal 8(375):ra41, 2015; Nature 550(7675):260-264, 2017). This chapter provides a detailed description of procedures and important considerations when performing the protocols for the respective assays used to determine the effects of Hippo signaling on tissue repair and regeneration.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Regenerative Medicine , Signal Transduction , Acetaminophen/adverse effects , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hepatectomy , Hippo Signaling Pathway , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Regeneration
10.
Oncogene ; 37(42): 5618-5632, 2018 10.
Article in English | MEDLINE | ID: mdl-29907771

ABSTRACT

miR-30 is a microRNA frequently overexpressed in human cancers. However, the biological consequence of miR-30 overexpression in cancer has been unclear. In a genetic screen, miR-30 was found to abrogate oncogenic-induced senescence, a key tumor-suppressing mechanism that involves DNA damage responses, activation of p53 and induction of p16INK4A. In cells and mouse models, miR-30 disrupts senescence and promotes cancer by suppressing 2 targets, CHD7 and TNRC6A. We show that while CHD7 is a transcriptional coactivator essential for induction of p16INK4A in senescent cells, TNRC6A, a miRNA machinery component, is required for expression and functionality of DNA damage response RNAs (DDRNAs) that mediate DNA damage responses and p53 activation by orchestrating histone modifications, chromatin remodeling and recruitment of DNA damage factors at damaged sites. Thus, miR-30 inhibits both p16INK4A and p53, 2 key senescence effectors, leading to efficient senescence disruption. These findings have identified novel signaling pathways mediating oncogene-induced senescence and tumor-suppression, and revealed the molecular and cellular mechanisms underlying the oncogenic activity of miR-30. Thus, the miR-30/CHD7/TNRC6A pathway is potentially a novel diagnostic biomarker and therapeutic target for cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage/genetics , MicroRNAs/physiology , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Mice, Transgenic
11.
Cell Mol Immunol ; 15(12): 1003-1009, 2018 12.
Article in English | MEDLINE | ID: mdl-29568120

ABSTRACT

The Hippo signaling pathway has been established as a key regulator of organ size control, tumor suppression, and tissue regeneration in multiple organisms. Recently, emerging evidence has indicated that Hippo signaling might play an important role in regulating the immune system in both Drosophila and mammals. In particular, patients bearing a loss-of-function mutation of MST1 are reported to have an autosomal recessive primary immunodeficiency syndrome. MST1/2 kinases, the mammalian orthologs of Drosophila Hippo, may activate the non-canonical Hippo signaling pathway via MOB1A/B and/or NDR1/2 or cross-talk with other essential signaling pathways to regulate both innate and adaptive immunity. In this review, we present and discuss recent findings of cellular mechanisms/functions of Hippo signaling in the innate immunity in Drosophila and in mammals, T cell immunity, as well as the implications of Hippo signaling for tumor immunity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/immunology , Immunologic Deficiency Syndromes/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Drosophila Proteins/genetics , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction
12.
Nat Immunol ; 19(9): 1036, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29449628

ABSTRACT

In the version of this article initially published, the institution name for affiliation 3 (Maryland Anderson Cancer Center) was incorrect. The correct institution is MD Anderson Cancer Center. The error has been corrected in the HTML and PDF versions of the article.

13.
Theranostics ; 8(22): 6121-6131, 2018.
Article in English | MEDLINE | ID: mdl-30613287

ABSTRACT

The detection of cardiac markers is critical to the diagnosis of acute myocardial infarction, and immunochromatographic assays are a common tool for point-of-care analysis. Methods: We report a multiplexed lateral flow test strip for simultaneous quantitative detection of cardiac troponin I (cTnI), creatine kinase isoenzyme MB (CKMB), and myoglobin (Myo). Hydrophilic, monodisperse, stable, and carboxyl-modified (COOH-) magnetic nanobeads (MNBs) were used to construct immunomagnetic probes specific to the three cardiac markers. The detection area of the sandwich-style complexes contained three test lines for cTnI, CKMB, and Myo. The magnetic signal intensity of the detection area in the nitrocellulose membrane was measured via a magnetic immunochromatography reader developed in house. Results: To optimize the assay, a modified working buffer was also investigated to improve the detection sensitivity, decrease the background noise, and shorten the detection time. The MNB-based immunochromatography test (MICT) strip offers a wide linear dynamic detection range, rapid detection, high sensitivity, and specificity. The limit of detection was 0.0089 ng/mL for cTnI, 0.063 ng/mL for CKMB, and 0.05 ng/mL for Myo with minimal cross-reactivity. There were 110 clinical human serum samples that were used to evaluate this platform with high correlation. Conclusion: MICT shows great potential as a supplemental method for in vitro diagnostics in the laboratory or in other point-of-care testing (POCT) applications.


Subject(s)
Biomarkers/blood , Immunoassay/methods , Magnetics/methods , Myocardial Infarction/diagnosis , Creatine Kinase, MB Form/blood , Humans , Immunoassay/instrumentation , Magnetics/instrumentation , Myocardial Infarction/blood , Myoglobin/blood , Nanostructures , Point-of-Care Systems , Sensitivity and Specificity , Troponin I/blood
15.
Nat Immunol ; 18(7): 800-812, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28504697

ABSTRACT

An imbalance in the lineages of immunosuppressive regulatory T cells (Treg cells) and the inflammatory TH17 subset of helper T cells leads to the development of autoimmune and/or inflammatory disease. Here we found that TAZ, a coactivator of TEAD transcription factors of Hippo signaling, was expressed under TH17 cell-inducing conditions and was required for TH17 differentiation and TH17 cell-mediated inflammatory diseases. TAZ was a critical co-activator of the TH17-defining transcription factor RORγt. In addition, TAZ attenuated Treg cell development by decreasing acetylation of the Treg cell master regulator Foxp3 mediated by the histone acetyltransferase Tip60, which targeted Foxp3 for proteasomal degradation. In contrast, under Treg cell-skewing conditions, TEAD1 expression and sequestration of TAZ from the transcription factors RORγt and Foxp3 promoted Treg cell differentiation. Furthermore, deficiency in TAZ or overexpression of TEAD1 induced Treg cell differentiation, whereas expression of a transgene encoding TAZ or activation of TAZ directed TH17 cell differentiation. Our results demonstrate a pivotal role for TAZ in regulating the differentiation of Treg cells and TH17 cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Differentiation/immunology , Colitis/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intracellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Arthritis, Rheumatoid/immunology , Case-Control Studies , Chromatin Immunoprecipitation , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Immunoblotting , Lysine Acetyltransferase 5 , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sjogren's Syndrome/immunology , Smad Proteins/immunology , Smad Proteins/metabolism , TEA Domain Transcription Factors , Trans-Activators/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins
16.
Cancer Cell ; 31(5): 669-684.e7, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28486106

ABSTRACT

Polyploidy can lead to aneuploidy and tumorigenesis. Here, we report that the Hippo pathway effector Yap promotes the diploid-polyploid conversion and polyploid cell growth through the Akt-Skp2 axis. Yap strongly induces the acetyltransferase p300-mediated acetylation of the E3 ligase Skp2 via Akt signaling. Acetylated Skp2 is exclusively localized to the cytosol, which causes hyper-accumulation of the cyclin-dependent kinase inhibitor p27, leading to mitotic arrest and subsequently cell polyploidy. In addition, the pro-apoptotic factors FoxO1/3 are overly degraded by acetylated Skp2, resulting in polyploid cell division, genomic instability, and oncogenesis. Importantly, the depletion or inactivation of Akt or Skp2 abrogated Hippo signal deficiency-induced liver tumorigenesis, indicating their epistatic interaction. Thus, we conclude that Hippo-Yap signaling suppresses cell polyploidy and oncogenesis through Skp2.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms/enzymology , Ploidies , Protein Serine-Threonine Kinases/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytosol/enzymology , Epistasis, Genetic , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Hep G2 Cells , Hippo Signaling Pathway , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Transgenic , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pregnancy , Protein Serine-Threonine Kinases/genetics , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , S-Phase Kinase-Associated Proteins/genetics , Signal Transduction , Time Factors , Transcription Factors , Transfection , YAP-Signaling Proteins , p300-CBP Transcription Factors/metabolism
17.
Sci Transl Med ; 8(352): 352ra108, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27535619

ABSTRACT

Tissue repair and regenerative medicine address the important medical needs to replace damaged tissue with functional tissue. Most regenerative medicine strategies have focused on delivering biomaterials and cells, yet there is the untapped potential for drug-induced regeneration with good specificity and safety profiles. The Hippo pathway is a key regulator of organ size and regeneration by inhibiting cell proliferation and promoting apoptosis. Kinases MST1 and MST2 (MST1/2), the mammalian Hippo orthologs, are central components of this pathway and are, therefore, strong target candidates for pharmacologically induced tissue regeneration. We report the discovery of a reversible and selective MST1/2 inhibitor, 4-((5,10-dimethyl-6-oxo-6,10-dihydro-5H-pyrimido[5,4-b]thieno[3,2-e][1,4]diazepin-2-yl)amino)benzenesulfonamide (XMU-MP-1), using an enzyme-linked immunosorbent assay-based high-throughput biochemical assay. The cocrystal structure and the structure-activity relationship confirmed that XMU-MP-1 is on-target to MST1/2. XMU-MP-1 blocked MST1/2 kinase activities, thereby activating the downstream effector Yes-associated protein and promoting cell growth. XMU-MP-1 displayed excellent in vivo pharmacokinetics and was able to augment mouse intestinal repair, as well as liver repair and regeneration, in both acute and chronic liver injury mouse models at a dose of 1 to 3 mg/kg via intraperitoneal injection. XMU-MP-1 treatment exhibited substantially greater repopulation rate of human hepatocytes in the Fah-deficient mouse model than in the vehicle-treated control, indicating that XMU-MP-1 treatment might facilitate human liver regeneration. Thus, the pharmacological modulation of MST1/2 kinase activities provides a novel approach to potentiate tissue repair and regeneration, with XMU-MP-1 as the first lead for the development of targeted regenerative therapeutics.


Subject(s)
Hepatocyte Growth Factor/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Regeneration/drug effects , Sulfonamides/pharmacology , Acetaminophen/toxicity , Animals , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/drug therapy , Colitis/chemically induced , Colitis/prevention & control , Crystallization , Hepatocyte Growth Factor/chemistry , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/transplantation , High-Throughput Screening Assays , Humans , Intracellular Signaling Peptides and Proteins , Lung Injury/drug therapy , Mice , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Regenerative Medicine , Serine-Threonine Kinase 3 , Signal Transduction/drug effects , Sulfonamides/chemistry , Translational Research, Biomedical
18.
Nat Commun ; 6: 8371, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26381214

ABSTRACT

The receptor-interacting protein kinase 3 (RIPK3) plays crucial roles in programmed necrosis and innate inflammatory responses. However, a little is known about the involvement of RIPK3 in NKT cell-mediated immune responses. Here, we demonstrate that RIPK3 plays an essential role in NKT cell function via activation of the mitochondrial phosphatase phosphoglycerate mutase 5 (PGAM5). RIPK3-mediated activation of PGAM5 promotes the expression of cytokines by facilitating nuclear translocation of NFAT and dephosphorylation of dynamin-related protein 1 (Drp1), a GTPase is essential for mitochondrial homoeostasis. Ripk3(-/-) mice show reduced NKT cell responses to metastatic tumour cells, and both deletion of RIPK3 and pharmacological inhibition of Drp1 protects mice from NKT cell-mediated induction of acute liver damage. Collectively, the results identify a crucial role for RIPK3-PGAM5-Drp1/NFAT signalling in NKT cell activation, and further suggest that RIPK3-PGAM5 signalling may mediate crosstalk between mitochondrial function and immune signalling.


Subject(s)
Dynamins/immunology , Immunity, Cellular/immunology , Liver/immunology , Natural Killer T-Cells/immunology , Phosphoric Monoester Hydrolases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Active Transport, Cell Nucleus , Animals , Blotting, Western , Cytokines/immunology , Dynamins/metabolism , HEK293 Cells , Hepatocytes , Humans , Inflammation , Interferon-gamma/immunology , Interleukin-4/immunology , Jurkat Cells , Melanoma, Experimental , Mice , Mice, Knockout , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neoplasm Transplantation , Phosphoprotein Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/immunology
19.
Nat Immunol ; 16(11): 1142-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26414765

ABSTRACT

Mitochondria need to be juxtaposed to phagosomes for the synergistic production of ample reactive oxygen species (ROS) in phagocytes to kill pathogens. However, how phagosomes transmit signals to recruit mitochondria has remained unclear. Here we found that the kinases Mst1 and Mst2 functioned to control ROS production by regulating mitochondrial trafficking and mitochondrion-phagosome juxtaposition. Mst1 and Mst2 activated the GTPase Rac to promote Toll-like receptor (TLR)-triggered assembly of the TRAF6-ECSIT complex that is required for the recruitment of mitochondria to phagosomes. Inactive forms of Rac, including the human Rac2(D57N) mutant, disrupted the TRAF6-ECSIT complex by sequestering TRAF6 and substantially diminished ROS production and enhanced susceptibility to bacterial infection. Our findings demonstrate that the TLR-Mst1-Mst2-Rac signaling axis is critical for effective phagosome-mitochondrion function and bactericidal activity.


Subject(s)
Phagocytes/immunology , Phagocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Infections/etiology , Bacterial Infections/immunology , Bacterial Infections/metabolism , Blood Bactericidal Activity/immunology , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/microbiology , Phagocytes/microbiology , Phagosomes/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Protein Kinase C-alpha/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Sepsis/etiology , Sepsis/immunology , Sepsis/metabolism , Serine-Threonine Kinase 3 , Signal Transduction , TNF Receptor-Associated Factor 6 , Toll-Like Receptors/metabolism , Ubiquitination , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
20.
Nat Commun ; 6: 6239, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25695629

ABSTRACT

The role of the unfolded protein response (UPR) in tissue homeostasis remains largely unknown. Here we find that loss of Mst1/2, the mammalian Hippo orthologues, or their regulator WW45, leads to a remarkably enlarged endoplasmic reticulum (ER) size-associated UPR. Intriguingly, attenuation of the UPR by tauroursodeoxycholic acid (TUDCA) diminishes Mst1/2 mutant-driven liver overgrowth and tumorigenesis by promoting nuclear exit and degradation of Hippo downstream effector Yap. Yap is required for UPR activity and ER expansion to alleviate ER stress. During the adaptive stage of the UPR, PERK kinase-eIF2α axis activates Yap, while prolonged ER stress-induced Hippo signalling triggers assembly of the GADD34/PP1 complex in a negative feedback loop to inhibit Yap and promote apoptosis. Significantly, the deregulation of UPR signals associated with Yap activation is found in a substantial fraction of human hepatocellular carcinoma (HCC). Thus, we conclude Yap integrates Hippo and UPR signalling to control liver size and tumorigenesis.


Subject(s)
Carcinogenesis/pathology , Liver Neoplasms/pathology , Liver/growth & development , Liver/pathology , Protein Serine-Threonine Kinases/metabolism , Taurochenodeoxycholic Acid/pharmacology , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Carcinogenesis/drug effects , Cell Cycle Proteins , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Hep G2 Cells , Hepatocyte Growth Factor/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/ultrastructure , Humans , Liver Neoplasms/enzymology , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Sequence Data , Mutation/genetics , Organ Size/drug effects , Phosphoproteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , Serine-Threonine Kinase 3 , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL