Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Cell ; 151(7): 1401-3, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260132

RESUMO

Two new studies reveal ways in which the Wnt pathway commandeers Hippo components for signaling. Azzolin et al. show how the Hippo transcription factor TAZ mediates Wnt signals, and Rosenbluh et al. show how ß-catenin and YAP1 form a kinase-regulated complex with transcription factor TBX5.

2.
Proteomics ; : e2300641, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38629187

RESUMO

Human embryonic stem cells (hESCs) resemble the pluripotent epiblast cells found in the early postimplantation human embryo and represent the "primed" state of pluripotency. One factor that helps primed pluripotent cells retain pluripotency and prepare genes for differentiation is the transcription factor TCF7L1, a member of a small family of proteins known as T cell factors/Lymphoid enhancer factors (TCF/LEF) that act as downstream components of the WNT signaling pathway. Transcriptional output of the WNT pathway is regulated, in part, by the activity of TCF/LEFs in conjunction with another component of the WNT pathway, ß-CATENIN. Because TCF7L1 plays an important role in regulating pluripotency, we began to characterize the protein complex associated with TCF7L1 when bound to chromatin in hESCs using rapid immunoprecipitation of endogenous proteins (RIME).  Data are available via ProteomeXchange with identifier PXD047582. These data identify known and new partners of TCF7L1 on chromatin and provide novel insights into how TCF7L1 and pluripotency itself might be regulated.

3.
J Proteome Res ; 20(9): 4452-4461, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34351778

RESUMO

Recent advances in sample preparation enable label-free mass spectrometry (MS)-based proteome profiling of small numbers of mammalian cells. However, specific devices are often required to downscale sample processing volume from the standard 50-200 µL to sub-µL for effective nanoproteomics, which greatly impedes the implementation of current nanoproteomics methods by the proteomics research community. Herein, we report a facile one-pot nanoproteomics method termed SOPs-MS (surfactant-assisted one-pot sample processing at the standard volume coupled with MS) for convenient robust proteome profiling of 50-1000 mammalian cells. Building upon our recent development of SOPs-MS for label-free single-cell proteomics at a low µL volume, we have systematically evaluated its processing volume at 10-200 µL using 100 human cells. The processing volume of 50 µL that is in the range of volume for standard proteomics sample preparation has been selected for easy sample handling with a benchtop micropipette. SOPs-MS allows for reliable label-free quantification of ∼1200-2700 protein groups from 50 to 1000 MCF10A cells. When applied to small subpopulations of mouse colon crypt cells, SOPs-MS has revealed protein signatures between distinct subpopulation cells with identification of ∼1500-2500 protein groups for each subpopulation. SOPs-MS may pave the way for routine deep proteome profiling of small numbers of cells and low-input samples.


Assuntos
Proteoma , Proteômica , Animais , Cromatografia Líquida , Perfilação da Expressão Gênica , Espectrometria de Massas , Camundongos
4.
Development ; 145(4)2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29361574

RESUMO

Human embryonic stem cells (hESCs) are exquisitely sensitive to WNT ligands, which rapidly cause differentiation. Therefore, hESC self-renewal requires robust mechanisms to keep the cells in a WNT inactive but responsive state. How they achieve this is largely unknown. We explored the role of transcriptional regulators of WNT signaling, the TCF/LEFs. As in mouse ESCs, TCF7L1 is the predominant family member expressed in hESCs. Genome-wide, it binds a gene cohort involved in primitive streak formation at gastrulation, including NODAL, BMP4 and WNT3 Comparing TCF7L1-bound sites with those bound by the WNT signaling effector ß-catenin indicates that TCF7L1 acts largely on the WNT signaling pathway. TCF7L1 overlaps less with the pluripotency regulators OCT4 and NANOG than in mouse ESCs. Gain- and loss-of-function studies indicate that TCF7L1 suppresses gene cohorts expressed in the primitive streak. Interestingly, we find that BMP4, another driver of hESC differentiation, downregulates TCF7L1, providing a mechanism of BMP and WNT pathway intersection. Together, our studies indicate that TCF7L1 plays a major role in maintaining hESC pluripotency, which has implications for human development during gastrulation.


Assuntos
Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Linha Primitiva/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Via de Sinalização Wnt/genética , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Linhagem da Célula , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Humanos , Imuno-Histoquímica , Análise em Microsséries , Reação em Cadeia da Polimerase
5.
Nat Immunol ; 10(9): 992-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19648923

RESUMO

The differentiation of activated CD4(+) T cells into the T helper type 1 (T(H)1) or T(H)2 fate is regulated by cytokines and the transcription factors T-bet and GATA-3. Whereas interleukin 12 (IL-12) produced by antigen-presenting cells initiates the T(H)1 fate, signals that initiate the T(H)2 fate are not completely characterized. Here we show that early GATA-3 expression, required for T(H)2 differentiation, was induced by T cell factor 1 (TCF-1) and its cofactor beta-catenin, mainly from the proximal Gata3 promoter upstream of exon 1b. This activity was induced after T cell antigen receptor (TCR) stimulation and was independent of IL-4 receptor signaling through the transcription factor STAT6. Furthermore, TCF-1 blocked T(H)1 fate by negatively regulating interferon-gamma (IFN-gamma) expression independently of beta-catenin. Thus, TCF-1 initiates T(H)2 differentiation of activated CD4(+) T cells by promoting GATA-3 expression and suppressing IFN-gamma expression.


Assuntos
Fator de Transcrição GATA3/genética , Interferon gama/biossíntese , Fator 1 de Transcrição de Linfócitos T/fisiologia , Células Th2/fisiologia , Animais , Diferenciação Celular , Interleucina-12/biossíntese , Interleucina-4/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores Notch/fisiologia , beta Catenina/fisiologia
6.
Haematologica ; 104(7): 1365-1377, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30630973

RESUMO

Canonical Wnt/ß-catenin signaling is frequently dysregulated in myeloid leukemias and is implicated in leukemogenesis. Nuclear-localized ß-catenin is indicative of active Wnt signaling and is frequently observed in acute myeloid leukemia (AML) patients; however, some patients exhibit little or no nuclear ß-catenin even where cytosolic ß-catenin is abundant. Control of the subcellular localization of ß-catenin therefore represents an additional mechanism regulating Wnt signaling in hematopoietic cells. To investigate the factors mediating the nuclear-localization of ß-catenin, we carried out the first nuclear/cytoplasmic proteomic analysis of the ß-catenin interactome in myeloid leukemia cells and identified putative novel ß-catenin interactors. Comparison of interacting factors between Wnt-responsive cells (high nuclear ß-catenin) versus Wnt-unresponsive cells (low nuclear ß-catenin) suggested the transcriptional partner, LEF-1, could direct the nuclear-localization of ß-catenin. The relative levels of nuclear LEF-1 and ß-catenin were tightly correlated in both cell lines and in primary AML blasts. Furthermore, LEF-1 knockdown perturbed ß-catenin nuclear-localization and transcriptional activation in Wnt-responsive cells. Conversely, LEF-1 overexpression was able to promote both nuclear-localization and ß-catenin-dependent transcriptional responses in previously Wnt-unresponsive cells. This is the first ß-catenin interactome study in hematopoietic cells and reveals LEF-1 as a mediator of nuclear ß- catenin level in human myeloid leukemia.


Assuntos
Núcleo Celular/metabolismo , Leucemia Mieloide Aguda/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Síndromes Mielodisplásicas/metabolismo , Proteoma/análise , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Fator 1 de Ligação ao Facilitador Linfoide/antagonistas & inibidores , Fator 1 de Ligação ao Facilitador Linfoide/genética , Síndromes Mielodisplásicas/patologia , Domínios e Motivos de Interação entre Proteínas , RNA Interferente Pequeno/genética , Ativação Transcricional , Células Tumorais Cultivadas , Proteína Wnt1/genética , beta Catenina/genética
7.
EMBO J ; 33(7): 719-31, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24596249

RESUMO

Wnt signaling activates target genes by promoting association of the co-activator ß-catenin with TCF/LEF transcription factors. In the absence of ß-catenin, target genes are silenced by TCF-mediated recruitment of TLE/Groucho proteins, but the molecular basis for TLE/TCF-dependent repression is unclear. We describe the unusual three-dimensional structure of the N-terminal Q domain of TLE1 that mediates tetramerization and binds to TCFs. We find that differences in repression potential of TCF/LEFs correlates with their affinities for TLE-Q, rather than direct competition between ß-catenin and TLE for TCFs as part of an activation-repression switch. Structure-based mutation of the TLE tetramer interface shows that dimers cannot mediate repression, even though they bind to TCFs with the same affinity as tetramers. Furthermore, the TLE Q tetramer, not the dimer, binds to chromatin, specifically to K20 methylated histone H4 tails, suggesting that the TCF/TLE tetramer complex promotes structural transitions of chromatin to mediate repression.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Modelos Moleculares , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células COS , Linhagem Celular , Chlorocebus aethiops , Proteínas Correpressoras , Cristalografia , Histonas/metabolismo , Humanos , Metilação , Camundongos , Modelos Estruturais , Mutação , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Fatores de Transcrição TCF/metabolismo , Ativação Transcricional , beta Catenina/metabolismo
8.
EMBO J ; 33(13): 1454-73, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24825347

RESUMO

Much of the mechanism by which Wnt signaling drives proliferation during oncogenesis is attributed to its regulation of the cell cycle. Here, we show how Wnt/ß-catenin signaling directs another hallmark of tumorigenesis, namely Warburg metabolism. Using biochemical assays and fluorescence lifetime imaging microscopy (FLIM) to probe metabolism in vitro and in living tumors, we observe that interference with Wnt signaling in colon cancer cells reduces glycolytic metabolism and results in small, poorly perfused tumors. We identify pyruvate dehydrogenase kinase 1 (PDK1) as an important direct target within a larger gene program for metabolism. PDK1 inhibits pyruvate flux to mitochondrial respiration and a rescue of its expression in Wnt-inhibited cancer cells rescues glycolysis as well as vessel growth in the tumor microenvironment. Thus, we identify an important mechanism by which Wnt-driven Warburg metabolism directs the use of glucose for cancer cell proliferation and links it to vessel delivery of oxygen and nutrients.


Assuntos
Neoplasias do Colo/metabolismo , Glucose/metabolismo , Glicólise , Neovascularização Patológica/metabolismo , Microambiente Tumoral , Via de Sinalização Wnt , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Glucose/genética , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Consumo de Oxigênio/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil
9.
Mol Syst Biol ; 13(2): 912, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28183841

RESUMO

Cell-intrinsic metabolic reprogramming is a hallmark of cancer that provides anabolic support to cell proliferation. How reprogramming influences tumor heterogeneity or drug sensitivities is not well understood. Here, we report a self-organizing spatial pattern of glycolysis in xenograft colon tumors where pyruvate dehydrogenase kinase (PDK1), a negative regulator of oxidative phosphorylation, is highly active in clusters of cells arranged in a spotted array. To understand this pattern, we developed a reaction-diffusion model that incorporates Wnt signaling, a pathway known to upregulate PDK1 and Warburg metabolism. Partial interference with Wnt alters the size and intensity of the spotted pattern in tumors and in the model. The model predicts that Wnt inhibition should trigger an increase in proteins that enhance the range of Wnt ligand diffusion. Not only was this prediction validated in xenograft tumors but similar patterns also emerge in radiochemotherapy-treated colorectal cancer. The model also predicts that inhibitors that target glycolysis or Wnt signaling in combination should synergize and be more effective than each treatment individually. We validated this prediction in 3D colon tumor spheroids.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regulação para Cima , Via de Sinalização Wnt , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Camundongos , Modelos Teóricos , Transplante de Neoplasias , Piruvato Desidrogenase Quinase de Transferência de Acetil
10.
Nucleic Acids Res ; 42(22): 13615-32, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25414359

RESUMO

LEF/TCFs direct the final step in Wnt/ß-catenin signalling by recruiting ß-catenin to genes for activation of transcription. Ancient, non-vertebrate TCFs contain two DNA binding domains, a High Mobility Group box for recognition of the Wnt Response Element (WRE; 5'-CTTTGWWS-3') and the C-clamp domain for recognition of the GC-rich Helper motif (5'-RCCGCC-3'). Two vertebrate TCFs (TCF-1/TCF7 and TCF-4/TCF7L2) use the C-clamp as an alternatively spliced domain to regulate cell-cycle progression, but how the C-clamp influences TCF binding and activity genome-wide is not known. Here, we used a doxycycline inducible system with ChIP-seq to assess how the C-clamp influences human TCF1 binding genome-wide. Metabolic pulse-labeling of nascent RNA with 4'Thiouridine was used with RNA-seq to connect binding to the Wnt transcriptome. We find that the C-clamp enables targeting to a greater number of gene loci for stronger occupancy and transcription regulation. The C-clamp uses Helper sites concurrently with WREs for gene targeting, but it also targets TCF1 to sites that do not have readily identifiable canonical WREs. The coupled ChIP-seq/4'Thiouridine-seq analysis identified new Wnt target genes, including additional regulators of cell proliferation. Thus, C-clamp containing isoforms of TCFs are potent transcriptional regulators with an expanded transcriptome directed by C-clamp-Helper site interactions.


Assuntos
Fator 1-alfa Nuclear de Hepatócito/metabolismo , Transcriptoma , Via de Sinalização Wnt , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Imunoprecipitação da Cromatina , DNA/química , DNA/metabolismo , Regulação da Expressão Gênica , Loci Gênicos , Fator 1-alfa Nuclear de Hepatócito/química , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Mutação , Motivos de Nucleotídeos , Ligação Proteica , Estrutura Terciária de Proteína , Elementos de Resposta , Análise de Sequência de DNA , Tiouridina
11.
J Biol Chem ; 289(26): 18122-36, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24841207

RESUMO

Secreted Wnt lipoproteins are cysteine-rich and lipid-modified morphogens that bind to the Frizzled (FZD) receptor and LDL receptor-related protein 6 (LRP6). Wnt engages FZD through protruding thumb and index finger domains, which are each assembled from paired ß strands secured by disulfide bonds and grasp two sides of the FZD ectodomain. The importance of Wnt disulfide bonds has been assumed but uncharacterized. We systematically analyzed cysteines and associated disulfide bonds in the prototypic Wnt3a. Our data show that mutation of any individual cysteine of Wnt3a results in covalent Wnt oligomers through ectopic intermolecular disulfide bond formation and diminishes/abolishes Wnt signaling. Although individual cysteine mutations in the amino part of the saposin-like domain and in the base of the index finger are better tolerated and permit residual Wnt3a secretion/activity, those in the amino terminus, the thumb, and at the tip of the index finger are incompatible with secretion and/or activity. A few select double cysteine mutants based on the disulfide bond pattern restore Wnt secretion/activity. Further, a double cysteine mutation at the index finger tip results in a Wnt3a with normal secretion but minimal FZD binding and dominant negative properties. Our results experimentally validate predictions from the Wnt crystal structure and highlight critical but different roles of the saposin-like and cytokine-like domains, including the thumb and the index finger in Wnt folding/secretion and FZD binding. Finally, we modified existing expression vectors for 19 epitope-tagged human WNT proteins by removal of a tag-supplied ectopic cysteine, thereby generating tagged WNT ligands active in canonical and non-canonical signaling.


Assuntos
Cisteína/química , Dissulfetos/química , Proteína Wnt3A/química , Proteína Wnt3A/metabolismo , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Cisteína/genética , Cisteína/metabolismo , Dissulfetos/metabolismo , Humanos , Ligantes , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência , Transdução de Sinais , Proteína Wnt3A/genética
12.
EMBO Rep ; 14(4): 347-55, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23449499

RESUMO

T-cell factor/lymphoid enhancer factor (TCF/LEF) proteins regulate transcription by recruiting ß-catenin and its associated co-regulators. Whether TCF/LEFs also recruit more factors through independent, direct interactions is not well understood. Here we discover Ring Finger Protein 14 (RNF14) as a new binding partner for all TCF/LEF transcription factors. We show that RNF14 positively regulates Wnt signalling in human cancer cells and in an in vivo zebrafish model by binding to target promoters with TCF and stabilizing ß-catenin recruitment. RNF14 depletion experiments demonstrate that it is crucial for colon cancer cell survival. Therefore, we have identified a key interacting factor of TCF/ß-catenin complexes to regulate Wnt gene transcription.


Assuntos
Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fatores de Transcrição TCF/metabolismo , Transcrição Gênica , beta Catenina/metabolismo , Animais , Sobrevivência Celular , Neoplasias do Colo , Técnicas de Silenciamento de Genes , Células HCT116 , Células HEK293 , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , RNA Interferente Pequeno/genética , Regulação para Cima , Via de Sinalização Wnt , Peixe-Zebra
14.
bioRxiv ; 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38712298

RESUMO

Several classification systems have been developed to define tumor subtypes in colorectal cancer (CRC). One system proposes that tumor heterogeneity derives in part from distinct cancer stem cell populations that co-exist as admixtures of varying proportions. However, the lack of single cell resolution has prohibited a definitive identification of these types of stem cells and therefore any understanding of how each influence tumor phenotypes. Here were report the isolation and characterization of two cancer stem cell subtypes from the SW480 CRC cell line. We find these cancer stem cells are oncogenic versions of the normal Crypt Base Columnar (CBC) and Regenerative Stem Cell (RSC) populations from intestinal crypts and that their gene signatures are consistent with the "Admixture" and other CRC classification systems. Using publicly available single cell RNA sequencing (scRNAseq) data from CRC patients, we determine that RSC and CBC cancer stem cells are commonly co-present in human CRC. To characterize influences on the tumor microenvironment, we develop subtype-specific xenograft models and we define their tumor microenvironments at high resolution via scRNAseq. RSCs create differentiated, inflammatory, slow growing tumors. CBCs create proliferative, undifferentiated, invasive tumors. With this enhanced resolution, we unify current CRC patient classification schema with TME phenotypes and organization.

15.
BMC Cell Biol ; 14: 44, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-24073846

RESUMO

BACKGROUND: A few reports suggested that low levels of Wnt signaling might drive cell reprogramming, but these studies could not establish a clear relationship between Wnt signaling and self-renewal networks. There are ongoing debates as to whether and how the Wnt/ß-catenin signaling is involved in the control of pluripotency gene networks. Additionally, whether physiological ß-catenin signaling generates stem-like cells through interactions with other pathways is as yet unclear. The nasopharyngeal carcinoma HONE1 cells have low expression of ß-catenin and wild-type expression of p53, which provided a possibility to study regulatory mechanism of stemness networks induced by physiological levels of Wnt signaling in these cells. RESULTS: Introduction of increased ß-catenin signaling, haploid expression of ß-catenin under control by its natural regulators in transferred chromosome 3, resulted in activation of Wnt/ß-catenin networks and dedifferentiation in HONE1 hybrid cell lines, but not in esophageal carcinoma SLMT1 hybrid cells that had high levels of endogenous ß-catenin expression. HONE1 hybrid cells displayed stem cell-like properties, including enhancement of CD24(+) and CD44(+) populations and generation of spheres that were not observed in parental HONE1 cells. Signaling cascades were detected in HONE1 hybrid cells, including activation of p53- and RB1-mediated tumor suppressor pathways, up-regulation of Nanog-, Oct4-, Sox2-, and Klf4-mediated pluripotency networks, and altered E-cadherin expression in both in vitro and in vivo assays. qPCR array analyses further revealed interactions of physiological Wnt/ß-catenin signaling with other pathways such as epithelial-mesenchymal transition, TGF-ß, Activin, BMPR, FGFR2, and LIFR- and IL6ST-mediated cell self-renewal networks. Using ß-catenin shRNA inhibitory assays, a dominant role for ß-catenin in these cellular network activities was observed. The expression of cell surface markers such as CD9, CD24, CD44, CD90, and CD133 in generated spheres was progressively up-regulated compared to HONE1 hybrid cells. Thirty-four up-regulated components of the Wnt pathway were identified in these spheres. CONCLUSIONS: Wnt/ß-catenin signaling regulates self-renewal networks and plays a central role in the control of pluripotency genes, tumor suppressive pathways and expression of cancer stem cell markers. This current study provides a novel platform to investigate the interaction of physiological Wnt/ß-catenin signaling with stemness transition networks.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Nasofaríngeas/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais/genética , beta Catenina/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Carcinoma , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal/genética , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Células Híbridas/metabolismo , Células Híbridas/patologia , Fator 4 Semelhante a Kruppel , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inibidores , beta Catenina/genética
16.
Mol Cell Proteomics ; 10(4): M110.007385, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21285413

RESUMO

Identification of proteins in RNA-protein complexes is an important step toward understanding regulation of RNA-based processes. Because of the lack of appropriate methodologies, many studies have relied on the creation of in vitro assembled RNA-protein complexes using synthetic RNA and cell extracts. Such complexes may not represent authentic RNPs as they exist in living cells as synthetic RNA may not fold properly and nonspecific RNA-protein interactions can form during cell lysis and purification processes. To circumvent limitations in current approaches, we have developed a novel integrated strategy namely MS2 in vivo biotin tagged RNA affinity purification (MS2-BioTRAP) to capture bona fide in vivo-assembled RNA-protein complexes. In this method, HB-tagged bacteriophage protein MS2 and stem-loop tagged target or control RNAs are co-expressed in cells. The tight association between MS2 and the RNA stem-loop tags allows efficient HB-tag based affinity purification of authentic RNA-protein complexes. Proteins associated with target RNAs are subsequently identified and quantified using SILAC-based quantitative mass spectrometry. Here the 1.2 kb internal ribosome entry site (IRES) from lymphoid enhancer factor-1 mRNA has been used as a proof-of-principle target RNA. An IRES target was chosen because of its importance in protein translation and our limited knowledge of proteins associated with IRES function. With a conventionally translated target RNA as control, 36 IRES binding proteins have been quantitatively identified including known IRES binding factors, novel interacting proteins, translation initiation factors (eIF4A-1, eIF-2A, and eIF3g), and ribosomal subunits with known noncanonical actions (RPS19, RPS7, and RPL26). Validation studies with the small molecule eIF4A-1 inhibitor Hippuristanol shows that translation of endogenous lymphoid enhancer factor-1 mRNA is especially sensitive to eIF4A-1 activity. Our work demonstrates that MS2 in vivo biotin tagged RNA affinity purification is an effective and versatile approach that is generally applicable for other RNA-protein complexes.


Assuntos
Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Proteoma/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regiões 5' não Traduzidas , Células HEK293 , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Ligação Proteica , Proteoma/isolamento & purificação , Proteômica/métodos , Proteínas de Ligação a RNA/isolamento & purificação , Espectrometria de Massas em Tandem
17.
Differentiation ; 84(2): 203-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22784633

RESUMO

Wnt ligands are secreted morphogens that control multiple developmental processes during embryogenesis and adult homeostasis. A diverse set of receptors and signals have been linked to individual Wnts, but the lack of tools for comparative analysis has limited the ability to determine which of these signals are general for the entire Wnt family, and which define subsets of differently acting ligands. We have created a versatile Gateway library of clones for all 19 human Wnts. An analysis comparing epitope-tagged and untagged versions of each ligand shows that despite their similar expression at the mRNA level, Wnts exhibit considerable variation in stability, processing and secretion. At least 14 out of the 19 Wnts activate ß-catenin-dependent signaling, an activity that is cell type-dependent and tracks with the stabilization of ß-catenin and LRP6 phosphorylation. We find that the core Wnt modification and secretion proteins Porcupine (PORCN) and Wntless (WLS) are essential for all Wnts to signal through ß-catenin-dependent and independent pathways. This comprehensive toolkit provides critical tools and new insights into human Wnt gene expression and function.


Assuntos
Biblioteca Gênica , Via Secretória , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Células 3T3 , Aciltransferases , Animais , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/genética
18.
Transl Res ; 255: 97-108, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36481562

RESUMO

Accurately modeling tumor biology and testing novel therapies on patient-derived cells is critically important to developing therapeutic regimens personalized to a patient's specific disease. The vascularized microtumor (VMT), or "tumor-on-a-chip," is a physiologic preclinical cancer model that incorporates key features of the native human tumor microenvironment within a transparent microfluidic platform, allowing rapid drug screening in vitro. Herein we optimize methods for generating patient-derived VMT (pVMT) using fresh colorectal cancer (CRC) biopsies and surgical resections to test drug sensitivities at the individual patient level. In response to standard chemotherapy and TGF-ßR1 inhibition, we observe heterogeneous responses between pVMT derived from 6 patient biopsies, with the pVMT recapitulating tumor growth, histological features, metabolic heterogeneity, and drug responses of actual CRC tumors. Our results suggest that a translational infrastructure providing rapid information from patient-derived tumor cells in the pVMT, as established in this study, will support efforts to improve patient outcomes.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/tratamento farmacológico , Microfluídica , Microambiente Tumoral
19.
Nucleic Acids Res ; 38(19): 6375-88, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20525792

RESUMO

Lymphoid enhancer factor 1 (LEF-1) mediates Wnt signaling via recruitment of ß-catenin to target genes. The LEF1 gene is aberrantly transcribed in colon cancers because promoter 1 (P1) is a Wnt target gene and is activated by TCF-ß-catenin complexes. A second promoter in intron 2 (P2) produces dominant negative LEF-1 isoforms (dnLEF-1), but P2 is silent because it is repressed by an upstream distal repressor element. In this study we identify Yin Yang 1 (YY1) transcription factor as the P2-specific factor necessary for repression. Site-directed mutagenesis and EMSA were used to identify a YY1-binding site at +25 in P2, and chromatin immunoprecipitation assays detected YY1 binding to endogenous LEF1 P2. Mutation of this site relieves P2 repression in transient transfections, and knockdown of endogenous YY1 relieves repression of integrated P2 reporter constructs and decreases the H3K9me3 epigenetic marks. YY1 is responsible for repressor specificity because introduction of a single YY1-binding site into the P1 promoter makes it sensitive to the distal repressor. We also show that induced expression of dnLEF-1 in colon cancer cells slows their rate of proliferation. We propose that YY1 plays an important role in preventing dnLEF-1 expression and growth inhibition in colon cancer.


Assuntos
Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Fator 1 de Ligação ao Facilitador Linfoide/genética , Proteínas Repressoras/metabolismo , Fator de Transcrição YY1/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Mutação , Regiões Promotoras Genéticas , Proteínas Repressoras/fisiologia , Fator de Transcrição YY1/fisiologia
20.
Life Sci Alliance ; 5(11)2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35831023

RESUMO

Crosstalk between the Hedgehog and MAPK signaling pathways occurs in several types of cancer and contributes to clinical resistance to Hedgehog pathway inhibitors. Here we show that MAP kinase-mediated phosphorylation weakens the binding of the GLI1 transcription factor to its negative regulator SUFU. ERK2 phosphorylates GLI1 on three evolutionarily conserved target sites (S102, S116, and S130) located near the high-affinity binding site for SUFU; these phosphorylations cooperate to weaken the affinity of GLI1-SUFU binding by over 25-fold. Phosphorylation of any one, or even any two, of the three sites does not result in the level of SUFU release seen when all three sites are phosphorylated. Tumor-derived mutations in R100 and S105, residues bordering S102, also diminish SUFU binding, collectively defining a novel evolutionarily conserved SUFU affinity-modulating region. In cultured mammalian cells, GLI1 variants containing phosphomimetic substitutions of S102, S116, and S130 displayed an increased ability to drive transcription. We conclude that multisite phosphorylation of GLI1 by ERK2 or other MAP kinases weakens GLI1-SUFU binding, thereby facilitating GLI1 activation and contributing to both physiological and pathological crosstalk.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno , Proteínas Repressoras , Proteína GLI1 em Dedos de Zinco , Animais , Sítios de Ligação , Células Cultivadas , Sequência Conservada , Proteínas Hedgehog/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação , Ligação Proteica , Proteína GLI1 em Dedos de Zinco/antagonistas & inibidores , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa