Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Nat Commun ; 14(1): 3907, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37400441

ABSTRACT

YAP is a key transcriptional co-activator of TEADs, it regulates cell growth and is frequently activated in cancer. In Malignant Pleural Mesothelioma (MPM), YAP is activated by loss-of-function mutations in upstream components of the Hippo pathway, while, in Uveal Melanoma (UM), YAP is activated in a Hippo-independent manner. To date, it is unclear if and how the different oncogenic lesions activating YAP impact its oncogenic program, which is particularly relevant for designing selective anti-cancer therapies. Here we show that, despite YAP being essential in both MPM and UM, its interaction with TEAD is unexpectedly dispensable in UM, limiting the applicability of TEAD inhibitors in this cancer type. Systematic functional interrogation of YAP regulatory elements in both cancer types reveals convergent regulation of broad oncogenic drivers in both MPM and UM, but also strikingly selective programs. Our work reveals unanticipated lineage-specific features of the YAP regulatory network that provide important insights to guide the design of tailored therapeutic strategies to inhibit YAP signaling across different cancer types.


Subject(s)
Adaptor Proteins, Signal Transducing , Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins , Epigenomics , Transcription Factors/genetics , Transcription Factors/metabolism , Signal Transduction/genetics
2.
ACS Chem Biol ; 17(6): 1401-1414, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35508359

ABSTRACT

Unbiased transcriptomic RNA-seq data has provided deep insights into biological processes. However, its impact in drug discovery has been narrow given high costs and low throughput. Proof-of-concept studies with Digital RNA with pertUrbation of Genes (DRUG)-seq demonstrated the potential to address this gap. We extended the DRUG-seq platform by subjecting it to rigorous testing and by adding an open-source analysis pipeline. The results demonstrate high reproducibility and ability to resolve the mechanism(s) of action for a diverse set of compounds. Furthermore, we demonstrate how this data can be incorporated into a drug discovery project aiming to develop therapeutics for schizophrenia using human stem cell-derived neurons. We identified both an on-target activation signature, induced by a set of chemically distinct positive allosteric modulators of the N-methyl-d-aspartate (NMDA) receptor, and independent off-target effects. Overall, the protocol and open-source analysis pipeline are a step toward industrializing RNA-seq for high-complexity transcriptomics studies performed at a saturating scale.


Subject(s)
Drug Discovery , Transcriptome , Drug Discovery/methods , Humans , RNA , Reproducibility of Results , Sequence Analysis, RNA/methods
3.
Nat Commun ; 13(1): 930, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177623

ABSTRACT

The Hippo/YAP pathway controls cell proliferation through sensing physical and spatial organization of cells. How cell-cell contact is sensed by Hippo signaling is poorly understood. Here, we identified the cell adhesion molecule KIRREL1 as an upstream positive regulator of the mammalian Hippo pathway. KIRREL1 physically interacts with SAV1 and recruits SAV1 to cell-cell contact sites. Consistent with the hypothesis that KIRREL1-mediated cell adhesion suppresses YAP activity, knockout of KIRREL1 increases YAP activity in neighboring cells. Analyzing pan-cancer CRISPR proliferation screen data reveals KIRREL1 as the top plasma membrane protein showing strong correlation with known Hippo regulators, highlighting a critical role of KIRREL1 in regulating Hippo signaling and cell proliferation. During liver regeneration in mice, KIRREL1 is upregulated, and its genetic ablation enhances hepatic YAP activity, hepatocyte reprogramming and biliary epithelial cell proliferation. Our data suggest that KIRREL1 functions as a feedback regulator of the mammalian Hippo pathway through sensing cell-cell interaction and recruiting SAV1 to cell-cell contact sites.


Subject(s)
Cell Communication , Cell Cycle Proteins/metabolism , Membrane Proteins/metabolism , Adult , Aged, 80 and over , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Feedback, Physiological , Female , Gene Knockout Techniques , HEK293 Cells , Hepatocytes , Hippo Signaling Pathway , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Middle Aged , YAP-Signaling Proteins/metabolism
4.
Commun Biol ; 4(1): 736, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34127790

ABSTRACT

Aggregates of hyperphosphorylated tau protein are a pathological hallmark of more than 20 distinct neurodegenerative diseases, including Alzheimer's disease, progressive supranuclear palsy, and frontotemporal dementia. While the exact mechanism of tau aggregation is unknown, the accumulation of aggregates correlates with disease progression. Here we report a genome-wide CRISPR screen to identify modulators of endogenous tau protein for the first time. Primary screens performed in SH-SY5Y cells, identified positive and negative regulators of tau protein levels. Hit validation of the top 43 candidate genes was performed using Ngn2-induced human cortical excitatory neurons. Using this approach, genes and pathways involved in modulation of endogenous tau levels were identified, including chromatin modifying enzymes, neddylation and ubiquitin pathway members, and components of the mTOR pathway. TSC1, a critical component of the mTOR pathway, was further validated in vivo, demonstrating the relevance of this screening strategy. These findings may have implications for treating neurodegenerative diseases in the future.


Subject(s)
Metabolic Networks and Pathways/genetics , Neurons/metabolism , tau Proteins/metabolism , Animals , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Cell Line, Tumor , Gene Editing , Genes/genetics , Genes/physiology , Genetic Testing/methods , Genome-Wide Association Study , Humans , Mice , Neuroblastoma/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism
5.
PLoS One ; 15(8): e0235551, 2020.
Article in English | MEDLINE | ID: mdl-32833964

ABSTRACT

VPS34 is a key regulator of endomembrane dynamics and cargo trafficking, and is essential in cultured cell lines and in mice. To better characterize the role of VPS34 in cell growth, we performed unbiased cell line profiling studies with the selective VPS34 inhibitor PIK-III and identified RKO as a VPS34-dependent cellular model. Pooled CRISPR screen in the presence of PIK-III revealed endolysosomal genes as genetic suppressors. Dissecting VPS34-dependent alterations with transcriptional profiling, we found the induction of hypoxia response and cholesterol biosynthesis as key signatures. Mechanistically, acute VPS34 inhibition enhanced lysosomal degradation of transferrin and low-density lipoprotein receptors leading to impaired iron and cholesterol uptake. Excess soluble iron, but not cholesterol, was sufficient to partially rescue the effects of VPS34 inhibition on mitochondrial respiration and cell growth, indicating that iron limitation is the primary driver of VPS34-dependency in RKO cells. Loss of RAB7A, an endolysosomal marker and top suppressor in our genetic screen, blocked transferrin receptor degradation, restored iron homeostasis and reversed the growth defect as well as metabolic alterations due to VPS34 inhibition. Altogether, our findings suggest that impaired iron mobilization via the VPS34-RAB7A axis drive VPS34-dependence in certain cancer cells.


Subject(s)
Class III Phosphatidylinositol 3-Kinases/metabolism , Iron/metabolism , Neoplasms/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Cholesterol/biosynthesis , Cholesterol/genetics , Class III Phosphatidylinositol 3-Kinases/genetics , Endosomes/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Receptors, LDL/metabolism , Transferrin/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
6.
Cell Rep ; 29(10): 2970-2978.e6, 2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31801065

ABSTRACT

A hallmark of chronic hepatitis B (CHB) virus infection is the presence of high circulating levels of non-infectious small lipid HBV surface antigen (HBsAg) vesicles. Although rare, sustained HBsAg loss is the idealized endpoint of any CHB therapy. A small molecule, RG7834, has been previously reported to inhibit HBsAg expression by targeting terminal nucleotidyltransferase proteins 4A and 4B (TENT4A and TENT4B). In this study, we describe a genome-wide CRISPR screen to identify other potential host factors required for HBsAg expression and to gain further insights into the mechanism of RG7834. We report more than 60 genes involved in regulating HBsAg and identify additional factors involved in RG7834 activity, including a zinc finger CCHC-type containing 14 (ZCCHC14) protein. We show that ZCCHC14, together with TENT4A/B, stabilizes HBsAg expression through HBV RNA tailing, providing a potential new therapeutic target to achieve functional cure in CHB patients.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/genetics , Host Microbial Interactions/genetics , Nuclear Proteins/genetics , Antigens, Surface/genetics , Antiviral Agents/pharmacology , Cell Line, Tumor , DNA, Viral/genetics , Genome-Wide Association Study/methods , Hep G2 Cells , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Host Microbial Interactions/drug effects , Humans , Polynucleotide Adenylyltransferase/genetics , Viral Load/drug effects , Viral Load/genetics
7.
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
8.
Nat Commun ; 10(1): 4676, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31611556

ABSTRACT

Resident adult epithelial stem cells maintain tissue homeostasis by balancing self-renewal and differentiation. The stem cell potential of human epidermal keratinocytes is retained in vitro but lost over time suggesting extrinsic and intrinsic regulation. Transcription factor-controlled regulatory circuitries govern cell identity, are sufficient to induce pluripotency and transdifferentiate cells. We investigate whether transcriptional circuitry also governs phenotypic changes within a given cell type by comparing human primary keratinocytes with intrinsically high versus low stem cell potential. Using integrated chromatin and transcriptional profiling, we implicate IRF2 as antagonistic to stemness and show that it binds and regulates active cis-regulatory elements at interferon response and antigen presentation genes. CRISPR-KD of IRF2 in keratinocytes with low stem cell potential increases self-renewal, migration and epidermis formation. These data demonstrate that transcription factor regulatory circuitries, in addition to maintaining cell identity, control plasticity within cell types and offer potential for therapeutic modulation of cell function.


Subject(s)
Interferon Regulatory Factor-2/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Humans , Interferon Regulatory Factor-2/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/physiology
9.
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
10.
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
11.
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
12.
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
13.
ACS Chem Biol ; 14(1): 20-26, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30461263

ABSTRACT

Using a comprehensive chemical genetics approach, we identified a member of the lignan natural product family, HTP-013, which exhibited significant cytotoxicity across various cancer cell lines. Correlation of compound activity across a panel of reporter gene assays suggested the vacuolar-type ATPase (v-ATPase) as a potential target for this compound. Additional cellular studies and a yeast haploinsufficiency screen strongly supported this finding. Competitive photoaffinity labeling experiments demonstrated that the ATP6V0A2 subunit of the v-ATPase complex binds directly to HTP-013, and further mutagenesis library screening identified resistance-conferring mutations in ATP6V0A2. The positions of these mutations suggest the molecule binds a novel pocket within the domain of the v-ATPase complex responsible for proton translocation. While other mechanisms of v-ATPase regulation have been described, such as dissociation of the complex or inhibition by natural products including bafilomycin A1 and concanamycin, this work provides detailed insight into a distinct binding pocket within the v-ATPase complex.


Subject(s)
Biological Products/metabolism , Biological Products/pharmacology , Vacuolar Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Binding Sites , Biological Products/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , HCT116 Cells , HEK293 Cells , Humans , Molecular Structure , Neurospora crassa/metabolism , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/chemistry
14.
Sci Rep ; 8(1): 16304, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389991

ABSTRACT

CRISPR/Cas9 mediated gene editing of patient-derived hematopoietic stem and progenitor cells (HSPCs) ex vivo followed by autologous transplantation of the edited HSPCs back to the patient can provide a potential cure for monogenic blood disorders such as ß-hemoglobinopathies. One challenge for this strategy is efficient delivery of the ribonucleoprotein (RNP) complex, consisting of purified Cas9 protein and guide RNA, into HSPCs. Because ß-hemoglobinopathies are most prevalent in developing countries, it is desirable to have a reliable, efficient, easy-to-use and cost effective delivery method. With this goal in mind, we developed TRansmembrane Internalization Assisted by Membrane Filtration (TRIAMF), a new method to quickly and effectively deliver RNPs into HSPCs by passing a RNP and cell mixture through a filter membrane. We achieved robust gene editing in HSPCs using TRIAMF and demonstrated that the multilineage colony forming capacities and the competence for engraftment in immunocompromised mice of HSPCs were preserved post TRIAMF treatment. TRIAMF is a custom designed system using inexpensive components and has the capacity to process HSPCs at clinical scale.


Subject(s)
Fetal Hemoglobin/genetics , Filtration/methods , Gene Editing/methods , Hematopoietic Stem Cell Transplantation , Ribonucleoproteins/genetics , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Electroporation/methods , Female , Fetal Hemoglobin/metabolism , Filtration/economics , Filtration/instrumentation , Genetic Therapy/economics , Genetic Therapy/instrumentation , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Humans , Membranes, Artificial , Mice , Models, Animal , RNA, Guide, Kinetoplastida/genetics , Transplantation, Autologous
15.
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
16.
Nat Methods ; 15(11): 941-946, 2018 11.
Article in English | MEDLINE | ID: mdl-30297964

ABSTRACT

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Gene Editing , Genome, Human , Hematopoietic Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL