Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Article in English | MEDLINE | ID: mdl-37937078

ABSTRACT

Introduction: Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematopoietic disorders characterized by ineffective hematopoiesis, cytopenias, and dysplasia. The gene encoding ten-eleven translocation 2 (tet2), a dioxygenase enzyme that catalyzes the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine, is a recurrently mutated tumor suppressor gene in MDS and other myeloid malignancies. Previously, we reported a stable zebrafish line with a loss-of-function mutation in the tet2 gene. The tet2m/m-mutant zebrafish developed a pre-MDS state with kidney marrow dysplasia, but normal circulating blood counts by 11 months of age and accompanying anemia, signifying the onset of MDS, by 24 months of age. Methods: In the current study, we collected progenitor cells from the kidney marrows of the adult tet2m/m and tet2wt/wt fish at 4 and 15 months of age and conducted enhanced reduced representation of bisulfite sequencing (ERRBS) and bulk RNA-seq to measure changes in DNA methylation and gene expression of hematopoietic stem and progenitor cells (HSPCs). Results and discussion: A global increase in DNA methylation of gene promoter regions and CpG islands was observed in tet2m/m HSPCs at 4 months of age when compared with the wild type. Furthermore, hypermethylated genes were significantly enriched for targets of SUZ12 and the metal-response-element-binding transcription factor 2 (MTF2)-involved in the polycomb repressive complex 2 (PRC2). However, between 4 and 15 months of age, we observed a paradoxical global decrease in DNA methylation in tet2m/m HSPCs. Gene expression analyses identified upregulation of genes associated with mTORC1 signaling and interferon gamma and alpha responses in tet2m/m HSPCs at 4 months of age when compared with the wild type. Downregulated genes in HSPCs of tet2-mutant fish at 4 months of age were enriched for cell cycle regulation, heme metabolism, and interleukin 2 (IL2)/signal transducer and activator of transcription 5 (STAT5) signaling, possibly related to increased self-renewal and clonal advantage in HSPCs with tet2 loss of function. Finally, there was an overall inverse correlation between overall increased promoter methylation and gene expression.

2.
Nature ; 622(7982): 393-401, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37821590

ABSTRACT

Recent human decedent model studies1,2 and compassionate xenograft use3 have explored the promise of porcine organs for human transplantation. To proceed to human studies, a clinically ready porcine donor must be engineered and its xenograft successfully tested in nonhuman primates. Here we describe the design, creation and long-term life-supporting function of kidney grafts from a genetically engineered porcine donor transplanted into a cynomolgus monkey model. The porcine donor was engineered to carry 69 genomic edits, eliminating glycan antigens, overexpressing human transgenes and inactivating porcine endogenous retroviruses. In vitro functional analyses showed that the edited kidney endothelial cells modulated inflammation to an extent that was indistinguishable from that of human endothelial cells, suggesting that these edited cells acquired a high level of human immune compatibility. When transplanted into cynomolgus monkeys, the kidneys with three glycan antigen knockouts alone experienced poor graft survival, whereas those with glycan antigen knockouts and human transgene expression demonstrated significantly longer survival time, suggesting the benefit of human transgene expression in vivo. These results show that preclinical studies of renal xenotransplantation could be successfully conducted in nonhuman primates and bring us closer to clinical trials of genetically engineered porcine renal grafts.


Subject(s)
Graft Rejection , Kidney Transplantation , Macaca fascicularis , Swine , Transplantation, Heterologous , Animals , Humans , Animals, Genetically Modified , Endothelial Cells/immunology , Endothelial Cells/metabolism , Graft Rejection/immunology , Graft Rejection/prevention & control , Kidney Transplantation/methods , Polysaccharides/deficiency , Swine/genetics , Transplantation, Heterologous/methods , Transgenes/genetics
3.
Immunity ; 54(5): 976-987.e7, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979589

ABSTRACT

Aerobic glycolysis-the Warburg effect-converts glucose to lactate via the enzyme lactate dehydrogenase A (LDHA) and is a metabolic feature of effector T cells. Cells generate ATP through various mechanisms and Warburg metabolism is comparatively an energy-inefficient glucose catabolism pathway. Here, we examined the effect of ATP generated via aerobic glycolysis in antigen-driven T cell responses. Cd4CreLdhafl/fl mice were resistant to Th17-cell-mediated experimental autoimmune encephalomyelitis and exhibited defective T cell activation, migration, proliferation, and differentiation. LDHA deficiency crippled cellular redox balance and inhibited ATP production, diminishing PI3K-dependent activation of Akt kinase and thereby phosphorylation-mediated inhibition of Foxo1, a transcriptional repressor of T cell activation programs. Th17-cell-specific expression of an Akt-insensitive Foxo1 recapitulated the defects seen in Cd4CreLdhafl/fl mice. Induction of LDHA required PI3K signaling and LDHA deficiency impaired PI3K-catalyzed PIP3 generation. Thus, Warburg metabolism augments glycolytic ATP production, fueling a PI3K-centered positive feedback regulatory circuit that drives effector T cell responses.


Subject(s)
Adenosine Triphosphate/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Signal Transduction/physiology , Th17 Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Female , Gene Expression Regulation, Neoplastic/physiology , Glucose/metabolism , Glycogen Storage Disease/metabolism , Glycolysis/physiology , L-Lactate Dehydrogenase/deficiency , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Nat Genet ; 52(2): 198-207, 2020 02.
Article in English | MEDLINE | ID: mdl-31932695

ABSTRACT

Mutations in ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, are the most common alterations of the SWI/SNF complex in estrogen-receptor-positive (ER+) breast cancer. We identify that ARID1A inactivating mutations are present at a high frequency in advanced endocrine-resistant ER+ breast cancer. An epigenome CRISPR-CAS9 knockout (KO) screen identifies ARID1A as the top candidate whose loss determines resistance to the ER degrader fulvestrant. ARID1A inactivation in cells and in patients leads to resistance to ER degraders by facilitating a switch from ER-dependent luminal cells to ER-independent basal-like cells. Cellular plasticity is mediated by loss of ARID1A-dependent SWI/SNF complex targeting to genomic sites of the luminal lineage-determining transcription factors including ER, forkhead box protein A1 (FOXA1) and GATA-binding factor 3 (GATA3). ARID1A also regulates genome-wide ER-FOXA1 chromatin interactions and ER-dependent transcription. Altogether, we uncover a critical role for ARID1A in maintaining luminal cell identity and endocrine therapeutic response in ER+ breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Mice , Mutation , Receptors, Estrogen/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
7.
Immunity ; 51(4): 766-779.e17, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31495665

ABSTRACT

Increasing evidence indicates CD4+ T cells can recognize cancer-specific antigens and control tumor growth. However, it remains difficult to predict the antigens that will be presented by human leukocyte antigen class II molecules (HLA-II), hindering efforts to optimally target them therapeutically. Obstacles include inaccurate peptide-binding prediction and unsolved complexities of the HLA-II pathway. To address these challenges, we developed an improved technology for discovering HLA-II binding motifs and conducted a comprehensive analysis of tumor ligandomes to learn processing rules relevant in the tumor microenvironment. We profiled >40 HLA-II alleles and showed that binding motifs were highly sensitive to HLA-DM, a peptide-loading chaperone. We also revealed that intratumoral HLA-II presentation was dominated by professional antigen-presenting cells (APCs) rather than cancer cells. Integrating these observations, we developed algorithms that accurately predicted APC ligandomes, including peptides from phagocytosed cancer cells. These tools and biological insights will enable improved HLA-II-directed cancer therapies.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Epitope Mapping/methods , HLA Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Immunotherapy/methods , Mass Spectrometry/methods , Neoplasms/therapy , Algorithms , Alleles , Antigen Presentation , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Datasets as Topic , HLA Antigens/genetics , HLA-D Antigens/metabolism , Humans , Neoplasms/immunology , Protein Binding , Protein Interaction Domains and Motifs/genetics , Software
8.
Nature ; 571(7765): 408-412, 2019 07.
Article in English | MEDLINE | ID: mdl-31243370

ABSTRACT

Mutations in the transcription factor FOXA1 define a unique subset of prostate cancers but the functional consequences of these mutations and whether they confer gain or loss of function is unknown1-9. Here, by annotating the landscape of FOXA1 mutations from 3,086 human prostate cancers, we define two hotspots in the forkhead domain: Wing2 (around 50% of all mutations) and the highly conserved DNA-contact residue R219 (around 5% of all mutations). Wing2 mutations are detected in adenocarcinomas at all stages, whereas R219 mutations are enriched in metastatic tumours with neuroendocrine histology. Interrogation of the biological properties of wild-type FOXA1 and fourteen FOXA1 mutants reveals gain of function in mouse prostate organoid proliferation assays. Twelve of these mutants, as well as wild-type FOXA1, promoted an exaggerated pro-luminal differentiation program, whereas two different R219 mutants blocked luminal differentiation and activated a mesenchymal and neuroendocrine transcriptional program. Assay for transposase-accessible chromatin using sequencing (ATAC-seq) of wild-type FOXA1 and representative Wing2 and R219 mutants revealed marked, mutant-specific changes in open chromatin at thousands of genomic loci and exposed sites of FOXA1 binding and associated increases in gene expression. Of note, ATAC-seq peaks in cells expressing R219 mutants lacked the canonical core FOXA1-binding motifs (GTAAAC/T) but were enriched for a related, non-canonical motif (GTAAAG/A), which was preferentially activated by R219-mutant FOXA1 in reporter assays. Thus, FOXA1 mutations alter its pioneering function and perturb normal luminal epithelial differentiation programs, providing further support for the role of lineage plasticity in cancer progression.


Subject(s)
Cell Differentiation/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Mutation , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Lineage , Chromatin/genetics , Chromatin/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/chemistry , Humans , Male , Mice , Mice, Inbred NOD , Nucleotide Motifs , Organoids/cytology , Organoids/metabolism
9.
Cell Rep ; 27(1): 294-306.e5, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943409

ABSTRACT

The PI3K pathway integrates extracellular stimuli to phosphorylate effectors such as AKT and serum-and-glucocorticoid-regulated kinase (SGK1). We have previously reported that the PI3K pathway regulates estrogen receptor (ER)-dependent transcription in breast cancer through the phosphorylation of the lysine methyltransferase KMT2D by AKT. Here, we show that PI3Kα inhibition, via a negative-feedback loop, activates SGK1 to promote chromatin-based regulation of ER-dependent transcription. PI3K/AKT inhibitors activate ER, which promotes SGK1 transcription through direct binding to its promoter. Elevated SGK1, in turn, phosphorylates KMT2D, suppressing its function, leading to a loss of methylation of lysine 4 on histone H3 (H3K4) and a repressive chromatin state at ER loci to attenuate ER activity. Thus, SGK1 regulates the chromatin landscape and ER-dependent transcription via the direct phosphorylation of KMT2D. These findings reveal an ER-SGK1-KMT2D signaling circuit aimed to attenuate ER response through a role for SGK1 to program chromatin and ER transcriptional output.


Subject(s)
Chromatin Assembly and Disassembly , Feedback, Physiological , Immediate-Early Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Estrogen/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Histones/metabolism , Humans , Immediate-Early Proteins/metabolism , MCF-7 Cells , Methylation , Neoplasm Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcriptional Activation
10.
Cell Stem Cell ; 24(1): 153-165.e7, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30472158

ABSTRACT

Leukemias exhibit a dysregulated developmental program mediated through both genetic and epigenetic mechanisms. Although IKZF2 is expressed in hematopoietic stem cells (HSCs), we found that it is dispensable for mouse and human HSC function. In contrast to its role as a tumor suppressor in hypodiploid B-acute lymphoblastic leukemia, we found that IKZF2 is required for myeloid leukemia. IKZF2 is highly expressed in leukemic stem cells (LSCs), and its deficiency results in defective LSC function. IKZF2 depletion in acute myeloid leukemia (AML) cells reduced colony formation, increased differentiation and apoptosis, and delayed leukemogenesis. Gene expression, chromatin accessibility, and direct IKZF2 binding in MLL-AF9 LSCs demonstrate that IKZF2 regulates a HOXA9 self-renewal gene expression program and inhibits a C/EBP-driven differentiation program. Ectopic HOXA9 expression and CEBPE depletion rescued the effects of IKZF2 depletion. Thus, our study shows that IKZF2 regulates the AML LSC program and provides a rationale to therapeutically target IKZF2 in myeloid leukemia.


Subject(s)
Cell Differentiation , Cell Self Renewal , DNA-Binding Proteins/physiology , Gene Expression Regulation, Leukemic , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Transcription Factors/physiology , Animals , Chromatin/genetics , Chromatin/metabolism , Female , Hematopoiesis , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/metabolism
11.
EMBO J ; 37(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30348863

ABSTRACT

The Hippo pathway and its nuclear effector Yap regulate organ size and cancer formation. While many modulators of Hippo activity have been identified, little is known about the Yap target genes that mediate these growth effects. Here, we show that yap-/- mutant zebrafish exhibit defects in hepatic progenitor potential and liver growth due to impaired glucose transport and nucleotide biosynthesis. Transcriptomic and metabolomic analyses reveal that Yap regulates expression of glucose transporter glut1, causing decreased glucose uptake and use for nucleotide biosynthesis in yap-/- mutants, and impaired glucose tolerance in adults. Nucleotide supplementation improves Yap deficiency phenotypes, indicating functional importance of glucose-fueled nucleotide biosynthesis. Yap-regulated glut1 expression and glucose uptake are conserved in mammals, suggesting that stimulation of anabolic glucose metabolism is an evolutionarily conserved mechanism by which the Hippo pathway controls organ growth. Together, our results reveal a central role for Hippo signaling in glucose metabolic homeostasis.


Subject(s)
Glucose/metabolism , Liver/embryology , Nucleotides/biosynthesis , Signal Transduction/physiology , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Glucose/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Mice , Nucleotides/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Serine-Threonine Kinase 3 , Trans-Activators/genetics , YAP-Signaling Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
12.
J Clin Invest ; 128(1): 294-308, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202482

ABSTRACT

Oncogenomic studies indicate that copy number variation (CNV) alters genes involved in tumor progression; however, identification of specific driver genes affected by CNV has been difficult, as these rearrangements are often contained in large chromosomal intervals among several bystander genes. Here, we addressed this problem and identified a CNV-targeted oncogene by performing comparative oncogenomics of human and zebrafish melanomas. We determined that the gene encoding growth differentiation factor 6 (GDF6), which is the ligand for the BMP family, is recurrently amplified and transcriptionally upregulated in melanoma. GDF6-induced BMP signaling maintained a trunk neural crest gene signature in melanomas. Additionally, GDF6 repressed the melanocyte differentiation gene MITF and the proapoptotic factor SOX9, thereby preventing differentiation, inhibiting cell death, and promoting tumor growth. GDF6 was specifically expressed in melanomas but not melanocytes. Moreover, GDF6 expression levels in melanomas were inversely correlated with patient survival. Our study has identified a fundamental role for GDF6 and BMP signaling in governing an embryonic cell gene signature to promote melanoma progression, thus providing potential opportunities for targeted therapy to treat GDF6-positive cancers.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Growth Differentiation Factor 6/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cell Line, Tumor , Female , Growth Differentiation Factor 6/genetics , HEK293 Cells , Humans , Ligands , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Proteins/genetics
13.
Nat Genet ; 49(6): 866-875, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28436985

ABSTRACT

The identity of the RNA-binding proteins (RBPs) that govern cancer stem cells remains poorly characterized. The MSI2 RBP is a central regulator of translation of cancer stem cell programs. Through proteomic analysis of the MSI2-interacting RBP network and functional shRNA screening, we identified 24 genes required for in vivo leukemia. Syncrip was the most differentially required gene between normal and myeloid leukemia cells. SYNCRIP depletion increased apoptosis and differentiation while delaying leukemogenesis. Gene expression profiling of SYNCRIP-depleted cells demonstrated a loss of the MLL and HOXA9 leukemia stem cell program. SYNCRIP and MSI2 interact indirectly though shared mRNA targets. SYNCRIP maintains HOXA9 translation, and MSI2 or HOXA9 overexpression rescued the effects of SYNCRIP depletion. Altogether, our data identify SYNCRIP as a new RBP that controls the myeloid leukemia stem cell program. We propose that targeting these RBP complexes might provide a novel therapeutic strategy in leukemia.


Subject(s)
Gene Expression Regulation, Leukemic , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Leukemia, Myeloid/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Survival , Female , Hematopoiesis/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/pathology , Leukemia, Myeloid/pathology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , RNA, Small Interfering , RNA-Binding Proteins/genetics , Xenograft Model Antitumor Assays
14.
Nat Biotechnol ; 35(4): 347-349, 2017 04.
Article in English | MEDLINE | ID: mdl-28263296

ABSTRACT

We present GuideScan software for the design of CRISPR guide RNA libraries that can be used to edit coding and noncoding genomic regions. GuideScan produces high-density sets of guide RNAs (gRNAs) for single- and paired-gRNA genome-wide screens. We also show that the trie data structure of GuideScan enables the design of gRNAs that are more specific than those designed by existing tools.


Subject(s)
Algorithms , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Silencing , Machine Learning , RNA, Small Interfering/genetics , Software , CRISPR-Cas Systems/genetics , Chromosome Mapping/methods , Sequence Analysis, RNA/methods
15.
BMC Bioinformatics ; 18(1): 153, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28259147

ABSTRACT

BACKGROUND: High-accuracy mass spectrometry enables near comprehensive quantification of the components of the cellular proteomes, increasingly including their chemically modified variants. Likewise, large-scale libraries of quantified synthetic peptides are becoming available, enabling absolute quantification of chemically modified proteoforms, and therefore systems-level analyses of changes of their absolute abundance and stoichiometry. Existing computational methods provide advanced tools for mass spectral analysis and statistical inference, but lack integrated functions for quantitative analysis of post-translationally modified proteins and their modification stoichiometry. RESULTS: Here, we develop ProteoModlR, a program for quantitative analysis of abundance and stoichiometry of post-translational chemical modifications across temporal and steady-state biological states. While ProteoModlR is intended for the analysis of experiments using isotopically labeled reference peptides for absolute quantitation, it also supports the analysis of labeled and label-free data, acquired in both data-dependent and data-independent modes for relative quantitation. Moreover, ProteoModlR enables functional analysis of sparsely sampled quantitative mass spectrometry experiments by inferring the missing values from the available measurements, without imputation. The implemented architecture includes parsing and normalization functions to control for common sources of technical variation. Finally, ProteoModlR's modular design and interchangeable format are optimally suited for integration with existing computational proteomics tools, thereby facilitating comprehensive quantitative analysis of cellular signaling. CONCLUSIONS: ProteoModlR and its documentation are available for download at http://github.com/kentsisresearchgroup/ProteoModlR as a stand-alone R package.


Subject(s)
Peptides/metabolism , Proteome/metabolism , Proteomics/methods , Evaluation Studies as Topic , Isotope Labeling , Mass Spectrometry , Protein Processing, Post-Translational , Software
16.
Elife ; 62017 03 28.
Article in English | MEDLINE | ID: mdl-28350298

ABSTRACT

Thyroid cancer is common, yet the sequence of alterations that promote tumor formation are incompletely understood. Here, we describe a novel model of thyroid carcinoma in zebrafish that reveals temporal changes due to BRAFV600E. Through the use of real-time in vivo imaging, we observe disruption in thyroid follicle structure that occurs early in thyroid development. Combinatorial treatment using BRAF and MEK inhibitors reversed the developmental effects induced by BRAFV600E. Adult zebrafish expressing BRAFV600E in thyrocytes developed invasive carcinoma. We identified a gene expression signature from zebrafish thyroid cancer that is predictive of disease-free survival in patients with papillary thyroid cancer. Gene expression studies nominated TWIST2 as a key effector downstream of BRAF. Using CRISPR/Cas9 to genetically inactivate a TWIST2 orthologue, we suppressed the effects of BRAFV600E and restored thyroid morphology and hormone synthesis. These data suggest that expression of TWIST2 plays a role in an early step of BRAFV600E-mediated transformation.


Subject(s)
Morphogenesis , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Gland/embryology , Thyroid Gland/physiology , Thyroid Hormones/metabolism , Thyroid Neoplasms/pathology , Twist-Related Protein 2/biosynthesis , Animals , Disease Models, Animal , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Proto-Oncogene Proteins B-raf/genetics , Twist-Related Protein 2/genetics , Zebrafish
17.
Science ; 354(6311): 481-484, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27708054

ABSTRACT

Aerobic glycolysis (the Warburg effect) is a metabolic hallmark of activated T cells and has been implicated in augmenting effector T cell responses, including expression of the proinflammatory cytokine interferon-γ (IFN-γ), via 3' untranslated region (3'UTR)-mediated mechanisms. Here, we show that lactate dehydrogenase A (LDHA) is induced in activated T cells to support aerobic glycolysis but promotes IFN-γ expression independently of its 3'UTR. Instead, LDHA maintains high concentrations of acetyl-coenzyme A to enhance histone acetylation and transcription of Ifng Ablation of LDHA in T cells protects mice from immunopathology triggered by excessive IFN-γ expression or deficiency of regulatory T cells. These findings reveal an epigenetic mechanism by which aerobic glycolysis promotes effector T cell differentiation and suggest that LDHA may be targeted therapeutically in autoinflammatory diseases.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Glycolysis/genetics , L-Lactate Dehydrogenase/metabolism , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/cytology , 3' Untranslated Regions , Acetylation , Aerobiosis , Animals , Gene Expression , Interferon-gamma/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5 , Lymphocyte Activation , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/cytology , Th1 Cells/enzymology
18.
Proc Natl Acad Sci U S A ; 113(38): E5562-71, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27588899

ABSTRACT

Selenium, an essential micronutrient known for its cancer prevention properties, is incorporated into a class of selenocysteine-containing proteins (selenoproteins). Selenoprotein H (SepH) is a recently identified nucleolar oxidoreductase whose function is not well understood. Here we report that seph is an essential gene regulating organ development in zebrafish. Metabolite profiling by targeted LC-MS/MS demonstrated that SepH deficiency impairs redox balance by reducing the levels of ascorbate and methionine, while increasing methionine sulfoxide. Transcriptome analysis revealed that SepH deficiency induces an inflammatory response and activates the p53 pathway. Consequently, loss of seph renders larvae susceptible to oxidative stress and DNA damage. Finally, we demonstrate that seph interacts with p53 deficiency in adulthood to accelerate gastrointestinal tumor development. Overall, our findings establish that seph regulates redox homeostasis and suppresses DNA damage. We hypothesize that SepH deficiency may contribute to the increased cancer risk observed in cohorts with low selenium levels.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/genetics , Gastrointestinal Neoplasms/genetics , Selenoproteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Female , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Male , Oxidation-Reduction , Oxidative Stress/genetics , Selenium/metabolism , Selenoproteins/metabolism , Transcriptome/genetics , Zebrafish/genetics
19.
Nat Cell Biol ; 18(8): 886-896, 2016 08.
Article in English | MEDLINE | ID: mdl-27428308

ABSTRACT

The Hippo pathway is an important regulator of organ size and tumorigenesis. It is unclear, however, how Hippo signalling provides the cellular building blocks required for rapid growth. Here, we demonstrate that transgenic zebrafish expressing an activated form of the Hippo pathway effector Yap1 (also known as YAP) develop enlarged livers and are prone to liver tumour formation. Transcriptomic and metabolomic profiling identify that Yap1 reprograms glutamine metabolism. Yap1 directly enhances glutamine synthetase (glul) expression and activity, elevating steady-state levels of glutamine and enhancing the relative isotopic enrichment of nitrogen during de novo purine and pyrimidine biosynthesis. Genetic or pharmacological inhibition of GLUL diminishes the isotopic enrichment of nitrogen into nucleotides, suppressing hepatomegaly and the growth of liver cancer cells. Consequently, Yap-driven liver growth is susceptible to nucleotide inhibition. Together, our findings demonstrate that Yap1 integrates the anabolic demands of tissue growth during development and tumorigenesis by reprogramming nitrogen metabolism to stimulate nucleotide biosynthesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Transformation, Neoplastic/genetics , Liver/growth & development , Phosphoproteins/genetics , Trans-Activators/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Glutamine/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phosphoproteins/metabolism , Transcription Factors , YAP-Signaling Proteins , Zebrafish
20.
Cell ; 164(3): 365-77, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26806130

ABSTRACT

Malignancy can be suppressed by the immune system in a process termed immunosurveillance. However, to what extent immunosurveillance occurs in spontaneous cancers and the composition of participating cell types remains obscure. Here, we show that cell transformation triggers a tissue-resident lymphocyte response in oncogene-induced murine cancer models. Non-circulating cytotoxic lymphocytes, derived from innate, T cell receptor (TCR)αß, and TCRγδ lineages, expand in early tumors. Characterized by high expression of NK1.1, CD49a, and CD103, these cells share a gene-expression signature distinct from those of conventional NK cells, T cells, and invariant NKT cells. Generation of these lymphocytes is dependent on the cytokine IL-15, but not the transcription factor Nfil3 that is required for the differentiation of tumor-infiltrating NK cells, and IL-15 deficiency, but not Nfil3 deficiency, results in accelerated tumor growth. These findings reveal a tumor-elicited immunosurveillance mechanism that engages unconventional type-1-like innate lymphoid cells and type 1 innate-like T cells.


Subject(s)
Lymphocytes/immunology , Mammary Neoplasms, Experimental/immunology , Monitoring, Immunologic , T-Lymphocyte Subsets/immunology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Granzymes/metabolism , Interleukin-15/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...