Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39091878

RESUMO

S-acyltransferases play integral roles in essential physiological processes including regulation of oncogenic signaling pathways. While discovered over 40 years ago the field still lacks specific S-acylation inhibitors thus the potential benefit of pharmacologically targeting S-acyltransferases for human disease is still unknown. Here we report the identification of an orally bioavailable acyltransferase inhibitor SD-066-4 that inhibits the acyltransferase ZDHHC20. We identified a specific alanine residue that accommodates the methyl group of SD-066-4, thus providing isoform selectivity. SD-066-4 stably reduces EGFR S-acylation in Kras mutant cells and blocks the growth of Kras mutant lung tumors extending overall survival. We find that lung cancer patients harboring deletions in ZDHHC20 or ZDHHC14 concurrent with Kras alterations have a significant survival benefit, underscoring the translational importance of these enzymes.

2.
Open Biol ; 11(10): 210033, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34610265

RESUMO

The epidermal growth factor receptor (EGFR) is an essential driver of oncogenic signalling, and EGFR inhibitors are some of the earliest examples of successful targeted therapies in multiple types of cancer. The tractability of EGFR as a therapeutic target is overshadowed by the inevitable drug resistance that develops. Overcoming resistance mechanisms requires a deeper understanding of EGFR regulation in cancer cells. In this review, we discuss our recent discovery that the palmitoyltransferase DHHC20 palmitoylates EGFR on the C-terminal domain and plays a critical role in signal regulation during oncogenesis. Inhibiting DHHC20 expression or mutating the palmitoylation site on EGFR alters the EGF-induced signalling kinetics from a transient signal to a sustained signal. The change in signalling is accompanied by a decrease in cell proliferation in multiple human cancer cell lines. Our in vivo studies demonstrate that ablating the gene Zdhhc20 by CRISPR/Cas9-mediated inhibition in a mouse model of oncogenic Kras-driven lung adenocarcinoma potently inhibits tumorigenesis. The negative effect on tumorigenesis is mediated by EGFR since the expression of a palmitoylation-resistant mutant form of EGFR also inhibits Kras-driven lung adenocarcinoma. Finally, reducing EGFR palmitoylation increases the sensitivity of multiple cancer cell lines to existing inhibitors of EGFR and downstream signalling effector pathways. We will discuss the implications of these effects and strategies for targeting these new vulnerabilities.


Assuntos
Aciltransferases/metabolismo , Adenocarcinoma de Pulmão/patologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Animais , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Lipoilação , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Mutação , Domínios Proteicos , Transdução de Sinais
3.
Sci Signal ; 13(621)2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127496

RESUMO

Non-small cell lung cancer (NSCLC) is often characterized by mutually exclusive mutations in the epidermal growth factor receptor (EGFR) or the guanosine triphosphatase KRAS. We hypothesized that blocking EGFR palmitoylation, previously shown to inhibit EGFR activity, might alter downstream signaling in the KRAS-mutant setting. Here, we found that blocking EGFR palmitoylation, by either knocking down the palmitoyltransferase DHHC20 or expressing a palmitoylation-resistant EGFR mutant, reduced activation of the kinase PI3K, the abundance of the transcription factor MYC, and the proliferation of cells in culture, as well as reduced tumor growth in a mouse model of KRAS-mutant lung adenocarcinoma. Knocking down DHHC20 reduced the growth of existing tumors derived from human KRAS-mutant lung cancer cells and increased the sensitivity of these cells to a PI3K inhibitor. Palmitoylated EGFR interacted with the PI3K regulatory subunit PIK3R1 (p85) and increased the recruitment of the PI3K heterodimer to the plasma membrane. Alternatively, blocking palmitoylation increased the association of EGFR with the MAPK adaptor Grb2 and decreased that with p85. This binary switching between MAPK and PI3K signaling, modulated by EGFR palmitoylation, was only observed in the presence of oncogenic KRAS. These findings suggest a mechanism whereby oncogenic KRAS saturates signaling through unpalmitoylated EGFR, reducing formation of the PI3K signaling complex. Future development of DHHC20 inhibitors to reduce EGFR-PI3K signaling could be beneficial to patients with KRAS-mutant tumors.


Assuntos
Carcinogênese/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Lipoilação , Neoplasias Pulmonares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Carcinogênese/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Pulmonares/genética , Camundongos , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas p21(ras)/genética
4.
Elife ; 72018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29648538

RESUMO

Wnt5a has been implicated in melanoma progression and metastasis, although the exact downstream signaling events that contribute to melanoma metastasis are poorly understood. Wnt5a signaling results in acyl protein thioesterase 1 (APT1) mediated depalmitoylation of pro-metastatic cell adhesion molecules CD44 and MCAM, resulting in increased melanoma invasion. The mechanistic details that underlie Wnt5a-mediated regulation of APT1 activity and cellular function remain unknown. Here, we show Wnt5a signaling regulates APT1 activity through induction of APT1 phosphorylation and we further investigate the functional role of APT1 phosphorylation on its depalmitoylating activity. We found phosphorylation increased APT1 depalmitoylating activity and reduced APT1 dimerization. We further determined APT1 phosphorylation increases melanoma invasion in vitro, and also correlated with increased tumor grade and metastasis. Our results further establish APT1 as an important regulator of melanoma invasion and metastatic behavior. Inhibition of APT1 may represent a novel way to treat Wnt5a driven cancers.


Assuntos
Movimento Celular , Melanoma/secundário , Processamento de Proteína Pós-Traducional , Tioléster Hidrolases/metabolismo , Proteína Wnt-5a/metabolismo , Antígeno CD146/genética , Antígeno CD146/metabolismo , Proliferação de Células , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Lipoilação , Melanoma/genética , Melanoma/metabolismo , Invasividade Neoplásica , Fosforilação , Conformação Proteica , Multimerização Proteica , Transdução de Sinais , Tioléster Hidrolases/química , Tioléster Hidrolases/genética , Células Tumorais Cultivadas , Proteína Wnt-5a/genética
5.
Sci Signal ; 11(511)2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29295957

RESUMO

Asymmetric cell division results in two distinctly fated daughter cells. A molecular hallmark of asymmetric division is the unequal partitioning of cell fate determinants. We have previously established that growth factor signaling promotes protein depalmitoylation to foster polarized protein localization, which, in turn, drives migration and metastasis. We report protein palmitoylation as a key mechanism for the asymmetric partitioning of the cell fate determinants Numb and ß-catenin through the activity of the depalmitoylating enzyme APT1. Using point mutations, we showed that specific palmitoylated residues on Numb were required for its asymmetric localization. By live-cell imaging, we showed that reciprocal interactions between APT1 and the Rho family GTPase CDC42 promoted the asymmetric localization of Numb and ß-catenin to the plasma membrane. This, in turn, restricted Notch- or Wnt-responsive transcriptional activity to one daughter cell. Moreover, we showed that altering APT1 abundance changed the transcriptional signatures of MDA-MB-231 triple receptor-negative breast cancer cells, similar to changes in Notch and ß-catenin-mediated Wnt signaling. We also showed that loss of APT1 depleted a specific subpopulation of tumorigenic cells in colony formation assays. Together, our findings suggest that APT1-mediated depalmitoylation is a major mechanism of asymmetric cell division that maintains Notch- and Wnt-associated protein dynamics, gene expression, and cellular functions.


Assuntos
Divisão Celular Assimétrica/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Notch/metabolismo , Tioléster Hidrolases/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , beta Catenina/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Feminino , Humanos , Lipoilação , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Mutação Puntual , Receptores Notch/genética , Tioléster Hidrolases/genética , Neoplasias de Mama Triplo Negativas/genética , Via de Sinalização Wnt , beta Catenina/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
6.
Biochem Biophys Res Commun ; 493(1): 213-219, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28899783

RESUMO

Currently, there are no effective therapeutic strategies targeting Kras driven cancers, and therefore, identifying new targeted therapies and overcoming drug resistance have become paramount for effective long-term cancer therapy. We have found that reducing expression of the palmitoyl transferase DHHC20 increases cell death induced by the EGFR inhibitor gefitinib in Kras and EGFR mutant cell lines, but not MCF7 cells harboring wildtype Kras. We show that the increased gefitinib sensitivity in cancer cells induced by DHHC20 inhibition is mediated directly through loss of palmitoylation on a previously identified cysteine residue in the C-terminal tail of EGFR. We utilized an EGFR point mutant in which the palmitoylated cysteine 1025 is mutated to alanine (EGFRC1025A), that results in receptor activation. Expression of the EGFR mutant alone in NIH3T3 cells does not increase sensitivity to gefitinib-induced cell death. However, when EGFRC1025A is expressed in cells expressing activated KrasG12V, EGFR inhibitor induced cell death is increased. Surprisingly, lung cancer cells harboring the EGFR inhibitor resistant mutation, T790M, become sensitive to EGFR inhibitor treatment when DHHC20 is inhibited. Finally, the small molecule, 2-bromopalmitate, which has been shown to inhibit palmitoyl transferases, acts synergistically with gefitinib to induce cell death in the gefitinib resistant cell line NCI-H1975.


Assuntos
Cisteína/genética , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Neoplasias Experimentais/fisiopatologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Cocarcinogênese , Cisteína/metabolismo , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Gefitinibe , Humanos , Lipoilação/efeitos dos fármacos , Lipoilação/genética , Células MCF-7 , Proteínas de Membrana , Mutação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Quinazolinas/administração & dosagem , Quinazolinas/farmacologia
7.
Methods Mol Biol ; 1481: 61-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27590152

RESUMO

The secreted ligand Wnt5a regulates cell polarity and polarized cell movement during development by signaling through the poorly defined noncanonical Wnt pathway. Cell polarity regulates most aspects of cell behavior including the organization of apical/basolateral membrane domains of epithelial cells, polarized cell divisions along a directional plane, and front rear polarity during cell migration. These characteristics of cell polarity allow coordinated cell movements required for tissue formation and organogenesis during embryonic development. Genetic model organisms have been used to identify multiple signaling pathways including Wnt5a that are required to establish cell polarity and regulate polarized cell behavior. However, the downstream signaling events that regulate these complex cellular processes are still poorly understood. The methods below describe assays to study Wnt5a-induced cell polarity in cultured cells, which may facilitate our understanding of these complex signaling pathways.


Assuntos
Polaridade Celular/genética , Proteína Wnt-5a/isolamento & purificação , Células Cultivadas , Desenvolvimento Embrionário/genética , Transdução de Sinais , Via de Sinalização Wnt , Proteína Wnt-5a/genética
8.
Mol Cell ; 62(3): 385-396, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153536

RESUMO

Inappropriate activation of the receptor tyrosine kinase EGFR contributes to a variety of human malignancies. Here we show a mechanism to induce vulnerability to an existing first line treatment for EGFR-driven cancers. We find that inhibiting the palmitoyltransferase DHHC20 creates a dependence on EGFR signaling for cancer cell survival. The loss of palmitoylation increases sustained EGFR signal activation and sensitizes cells to EGFR tyrosine kinase inhibition. Our work shows that the reversible modification of EGFR with palmitate "pins" the unstructured C-terminal tail to the plasma membrane, impeding EGFR activation. We identify by mass spectrometry palmitoylated cysteine residues within the C-terminal tail where mutation of the cysteine residues to alanine is sufficient to activate EGFR signaling promoting cell migration and transformation. Our results reveal that the targeting of a peripheral modulator of EGFR signaling, DHHC20, causes a loss of signal regulation and susceptibility to EGFR inhibitor-induced cell death.


Assuntos
Aciltransferases/metabolismo , Neoplasias da Mama/enzimologia , Receptores ErbB/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Aciltransferases/genética , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Morte Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisteína , Endocitose , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/química , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/genética , Proteína Adaptadora GRB2/metabolismo , Gefitinibe , Células HEK293 , Humanos , Lipoilação , Espectrometria de Massas , Camundongos , Mutação , Células NIH 3T3 , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Proteólise , Quinazolinas/farmacologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Transfecção
9.
Cancer Biol Ther ; 17(1): 114-22, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26560942

RESUMO

Menin is encoded by the MEN1 gene, which is mutated in an inherited human syndrome, multiple endocrine neoplasia type 1(MEN1). Menin is primarily nuclear protein, acting as a tumor suppressor in endocrine organs, but as an oncogenic factor in the mixed lineage leukemia, in a tissue-specific manner. Recently, the crystal structures of menin with different binding partners reveal menin as a key scaffold protein that functionally interacts with various partners to regulate gene transcription in the nucleus. However, outside the nucleus, menin also regulates multiple signaling pathways that traverse the cell surface membrane. The precise nature regarding to how menin associates with the membrane fraction is poorly understood. Here we show that a small fraction of menin associates with the cell membrane fraction likely via serine palmitoylation. Moreover, the majority of the membrane-associated menin may reside inside membrane vesicles, as menin is protected from trypsin-mediated proteolysis, but disruption of the membrane fraction using detergent abolishes the detection. Consistently, cellular staining for menin also reveals the distribution of menin in the cell membrane and the punctate-like cell organelles. Our findings suggest that part of intracellular menin associates with the cell membrane peripherally as well as resides within the membrane vesicles.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Membrana Celular/genética , Núcleo Celular/genética , Cristalografia por Raios X , Humanos , Lipoilação , Camundongos , Proteínas Associadas à Matriz Nuclear/química , Proteínas Associadas à Matriz Nuclear/genética , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Ratos , Serina/metabolismo , Transdução de Sinais/genética
10.
PLoS One ; 10(7): e0132437, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26200654

RESUMO

Cell competition is a form of cell-cell interaction by which cells compare relative levels of fitness, resulting in the active elimination of less-fit cells, "losers," by more-fit cells, "winners." Here, we show that in three routinely-used mammalian cell lines - U2OS, 3T3, and MDCK cells - sub-clones arise stochastically that exhibit context-dependent competitive behavior. Specifically, cell death is elicited when winner and loser sub-clones are cultured together but not alone. Cell competition and elimination in these cell lines is caspase-dependent and requires cell-cell contact but does not require de novo RNA synthesis. Moreover, we show that the phenomenon involves differences in cellular metabolism. Hence, our study demonstrates that cell competition is a common feature of immortalized mammalian cells in vitro and implicates cellular metabolism as a mechanism by which cells sense relative levels of "fitness."


Assuntos
Caspases/metabolismo , Comunicação Celular , Técnicas de Cocultura/métodos , Células 3T3 , Animais , Apoptose , Linhagem Celular , Cães , Humanos , Células Madin Darby de Rim Canino , Camundongos , RNA/metabolismo
11.
J Biol Chem ; 290(25): 15707-15716, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25944911

RESUMO

Wnt5a signaling regulates polarized cell behavior, but the downstream signaling events that promote cell polarity are not well understood. Our results show that Wnt5a promotes depalmitoylation of the melanoma cell adhesion molecule (MCAM) at cysteine 590. Mutation of Cys-590 to glycine is sufficient to polarize MCAM localization, similar to what is observed with Wnt5a stimulation. Inhibition of the depalmitoylating enzyme APT1 blocks Wnt5a-induced depalmitoylation, asymmetric MCAM localization, and cell invasion. Directly altering expression of the basal protein palmitoylation machinery is sufficient to promote cell invasion. Additionally, cancer mutations in palmitoyltransferases decrease MCAM palmitoylation and have impaired ability to suppress cell invasion. Our results provide evidence that Wnt5a induces protein depalmitoylation, which promotes polarized protein localization and cell invasion.


Assuntos
Lipoilação , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Tioléster Hidrolases/metabolismo , Proteínas Wnt/metabolismo , Antígeno CD146/biossíntese , Antígeno CD146/genética , Linhagem Celular Tumoral , Humanos , Mutação , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/patologia , Proteínas Proto-Oncogênicas/genética , Tioléster Hidrolases/genética , Proteínas Wnt/genética , Proteína Wnt-5a
12.
PLoS One ; 8(12): e81689, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349113

RESUMO

Here, we describe an engineering approach to quantitatively compare migration, morphologies, and adhesion for tumorigenic human fibrosarcoma cells (HT-1080s) and primary human dermal fibroblasts (hDFs) with the aim of identifying distinguishing properties of the transformed phenotype. Relative adhesiveness was quantified using self-assembled monolayer (SAM) arrays and proteolytic 3-dimensional (3D) migration was investigated using matrix metalloproteinase (MMP)-degradable poly(ethylene glycol) (PEG) hydrogels ("synthetic extracellular matrix" or "synthetic ECM"). In synthetic ECM, hDFs were characterized by vinculin-containing features on the tips of protrusions, multipolar morphologies, and organized actomyosin filaments. In contrast, HT-1080s were characterized by diffuse vinculin expression, pronounced ß1-integrin on the tips of protrusions, a cortically-organized F-actin cytoskeleton, and quantitatively more rounded morphologies, decreased adhesiveness, and increased directional motility compared to hDFs. Further, HT-1080s were characterized by contractility-dependent motility, pronounced blebbing, and cortical contraction waves or constriction rings, while quantified 3D motility was similar in matrices with a wide range of biochemical and biophysical properties (including collagen) despite substantial morphological changes. While HT-1080s were distinct from hDFs for each of the 2D and 3D properties investigated, several features were similar to WM239a melanoma cells, including rounded, proteolytic migration modes, cortical F-actin organization, and prominent uropod-like structures enriched with ß1-integrin, F-actin, and melanoma cell adhesion molecule (MCAM/CD146/MUC18). Importantly, many of the features observed for HT-1080s were analogous to cellular changes induced by transformation, including cell rounding, a disorganized F-actin cytoskeleton, altered organization of focal adhesion proteins, and a weakly adherent phenotype. Based on our results, we propose that HT-1080s migrate in synthetic ECM with functional properties that are a direct consequence of their transformed phenotype.


Assuntos
Movimento Celular/genética , Transformação Celular Neoplásica , Fibroblastos/patologia , Fenótipo , Actinas/genética , Actinas/metabolismo , Antígeno CD146/genética , Antígeno CD146/metabolismo , Adesão Celular , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Matriz Extracelular/química , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Hidrogéis , Integrina beta1/genética , Integrina beta1/metabolismo , Metaloproteinases da Matriz/química , Mimetismo Molecular , Cultura Primária de Células , Vinculina/genética , Vinculina/metabolismo
13.
Dev Cell ; 26(6): 645-57, 2013 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-24091015

RESUMO

Wnt5a directs the assembly of the Wnt-receptor-actin-myosin-polarity (WRAMP) structure, which integrates cell-adhesion receptors with F-actin and myosin to form a microfilament array associated with multivesicular bodies (MVBs). The WRAMP structure is polarized to the cell posterior, where it directs tail-end membrane retraction, driving forward translocation of the cell body. Here we define constituents of the WRAMP proteome, including regulators of microfilament and microtubule dynamics, protein interactions, and enzymatic activity. IQGAP1, a scaffold for F-actin nucleation and crosslinking, is necessary for WRAMP structure formation, potentially bridging microfilaments and MVBs. Vesicle coat proteins, including coatomer-I subunits, localize to and are required for the WRAMP structure. Electron microscopy and live imaging demonstrate movement of the ER to the WRAMP structure and plasma membrane, followed by elevation of intracellular Ca2+. Thus, Wnt5a controls directional movement by recruiting cortical ER to mobilize a rear-directed, localized Ca2+ signal, activating actomyosin contraction and adhesion disassembly for membrane retraction.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt/metabolismo , Actinas/metabolismo , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Polaridade Celular , Proteína Coatomer/metabolismo , Retículo Endoplasmático/ultraestrutura , Humanos , Microtúbulos/metabolismo , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Miosinas/metabolismo , Receptores Wnt/metabolismo , Proteína Wnt-5a , Proteínas Ativadoras de ras GTPase/metabolismo
14.
Mol Cell Biol ; 33(19): 3864-78, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23897429

RESUMO

During adipocyte differentiation, significant epigenomic changes occur in association with the implementation of the adipogenic program. We have previously shown that histone acetylation increases during differentiation in a manner dependent on acetyl coenzyme A (acetyl-CoA) production by the enzyme ATP-citrate lyase (ACL). Whether ACL regulates nuclear targets in addition to histones during differentiation is not clear. In this study, we report that DNA methyltransferase 1 (DNMT1) levels in adipocytes are controlled in part by ACL and that silencing of DNMT1 can accelerate adipocyte differentiation. DNMT1 gene expression is induced early in 3T3-L1 adipocyte differentiation during mitotic clonal expansion and is critical for maintenance of DNA and histone H3K9 methylation patterns during this period. In the absence of DNMT1, adipocyte-specific gene expression and lipid accumulation occur precociously. Later in differentiation, DNMT1 levels decline in an ACL-dependent manner. ACL-mediated suppression of DNMT1 occurs at least in part by promoting expression of microRNA 148a (miR-148a), which represses DNMT1. Ectopic expression of miR-148a accelerates differentiation under standard conditions and can partially rescue a hypermethylation-mediated differentiation block. The data suggest a role for DNMT1 in modulating the timing of differentiation and describe a novel ACL-miR-148a-dependent mechanism for regulating DNMT1 during adipogenesis.


Assuntos
ATP Citrato (pro-S)-Liase/genética , Adipócitos/metabolismo , Diferenciação Celular/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Células 3T3-L1 , ATP Citrato (pro-S)-Liase/metabolismo , Adipócitos/citologia , Animais , Western Blotting , Linhagem Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Expressão Gênica , Células HEK293 , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Mitose/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Mol Pharmacol ; 83(3): 594-604, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23229510

RESUMO

The Integrated Stress Response (ISR) is a signaling program that enables cellular adaptation to stressful conditions like hypoxia and nutrient deprivation in the tumor microenvironment. An important effector of the ISR is activating transcription factor 4 (ATF4), a transcription factor that regulates genes involved in redox homeostasis and amino acid metabolism and transport. Because both inhibition and overactivation of the ISR can induce tumor cell death, modulators of ATF4 expression could prove to be clinically useful. In this study, chemical libraries were screened for modulators of ATF4 expression. We identified one compound, E235 (N-(1-benzyl-piperidin-4-yl)-2-(4-fluoro-phenyl)-benzo[d]imidazo[2,1-b]thiazole-7-carboxamide), that activated the ISR and dose-dependently increased levels of ATF4 in transformed cells. A dose-dependent decrease in viability was observed in several mouse and human tumor cell lines, and knockdown of ATF4 significantly increased the antiproliferative effects of E235. Interestingly, low µM doses of E235 induced senescence in many cell types, including HT1080 human fibrosarcoma and B16F10 mouse melanoma cells. E235-mediated induction of senescence was not dependent on p21 or p53; however, p21 conferred protection against the growth inhibitory effects of E235. Treatment with E235 resulted in an increase in cells arrested at the G2/M phase with a concurrent decrease in S-phase cells. E235 also activated DNA damage response signaling, resulting in increased levels of Ser15-phosphorylated p53, γ-H2AX, and phosphorylated checkpoint kinase 2 (Chk2), although E235 does not appear to cause physical DNA damage. Induction of γ-H2AX was abrogated in ATF4 knockdown cells. Together, these results suggest that modulation of the ISR pathway with the small molecule E235 could be a promising antitumor strategy.


Assuntos
Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Feminino , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Humanos , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
16.
Mol Cell Biol ; 32(12): 2268-78, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22493067

RESUMO

The endoplasmic reticulum (ER) resident PKR-like kinase (PERK) is necessary for Akt activation in response to ER stress. We demonstrate that PERK harbors intrinsic lipid kinase, favoring diacylglycerol (DAG) as a substrate and generating phosphatidic acid (PA). This activity of PERK correlates with activation of mTOR and phosphorylation of Akt on Ser473. PERK lipid kinase activity is regulated in a phosphatidylinositol 3-kinase (PI3K) p85α-dependent manner. Moreover, PERK activity is essential during adipocyte differentiation. Because PA and Akt regulate many cellular functions, including cellular survival, proliferation, migratory responses, and metabolic adaptation, our findings suggest that PERK has a more extensive role in insulin signaling, insulin resistance, obesity, and tumorigenesis than previously thought.


Assuntos
Adipócitos/enzimologia , Diferenciação Celular , eIF-2 Quinase/metabolismo , Adipócitos/citologia , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Ativação Enzimática , Metabolismo dos Lipídeos , Camundongos , Ácidos Fosfatídicos/metabolismo , Fosfatidilinositol 3-Quinases , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
17.
Mol Cell Proteomics ; 8(10): 2308-20, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19620624

RESUMO

RhoA controls changes in cell morphology and invasion associated with cancer phenotypes. Cell lines derived from melanoma tumors at varying stages revealed that RhoA is selectively activated in cells of metastatic origin. We describe a functional proteomics strategy to identify proteins regulated by RhoA and report a previously uncharacterized human protein, named "mediator of RhoA-dependent invasion (MRDI)," that is induced in metastatic cells by constitutive RhoA activation and promotes cell invasion. In human melanomas, MRDI localization correlated with stage, showing nuclear localization in nevi and early stage tumors and cytoplasmic localization with plasma membrane accentuation in late stage tumors. Consistent with its role in promoting cell invasion, MRDI localized to cell protrusions and leading edge membranes in cultured cells and was required for cell motility, tyrosine phosphorylation of focal adhesion kinase, and modulation of actin stress fibers. Unexpectedly MRDI had enzymatic function as an isomerase that converts the S-adenosylmethionine catabolite 5-methylribose 1-phosphate into 5-methylribulose 1-phosphate. The enzymatic function of MRDI was required for methionine salvage from S-adenosylmethionine but distinct from its function in cell invasion. Thus, mechanisms used by signal transduction pathways to control cell movement have evolved from proteins with ancient function in amino acid metabolism.


Assuntos
Aldose-Cetose Isomerases/metabolismo , Melanoma , Metionina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Aldose-Cetose Isomerases/genética , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Feminino , Humanos , Melanoma/enzimologia , Melanoma/patologia , Metionina/química , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Estrutura Molecular , Invasividade Neoplásica , Metástase Neoplásica , Proteômica/métodos , Interferência de RNA , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Transdução de Sinais/fisiologia , Transplante Heterólogo , Proteína rhoA de Ligação ao GTP/genética
18.
Mol Cell ; 34(1): 115-31, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19362540

RESUMO

Melanoma and other cancers harbor oncogenic mutations in the protein kinase B-Raf, which leads to constitutive activation and dysregulation of MAP kinase signaling. In order to elucidate molecular determinants responsible for B-Raf control of cancer phenotypes, we present a method for phosphoprotein profiling, using negative ionization mass spectrometry to detect phosphopeptides based on their fragment ion signature caused by release of PO(3)(-). The method provides an alternative strategy for phosphoproteomics, circumventing affinity enrichment of phosphopeptides and isotopic labeling of samples. Ninety phosphorylation events were regulated by oncogenic B-Raf signaling, based on their responses to treating melanoma cells with MKK1/2 inhibitor. Regulated phosphoproteins included known signaling effectors and cytoskeletal regulators. We investigated MINERVA/FAM129B, a target belonging to a protein family with unknown category and function, and established the importance of this protein and its MAP kinase-dependent phosphorylation in controlling melanoma cell invasion into three-dimensional collagen matrix.


Assuntos
Melanoma/metabolismo , Proteômica , Proteínas Proto-Oncogênicas B-raf/metabolismo , Linhagem Celular Tumoral , Humanos , Sistema de Sinalização das MAP Quinases , Espectrometria de Massas , Mutação , Fosfoproteínas/análise , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/fisiologia , Especificidade por Substrato
19.
Dev Biol ; 327(1): 12-23, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19084000

RESUMO

The endoderm of C. elegans arises entirely from a single progenitor cell, the E blastomere, whose identity is specified by GATA type transcription factors, including END-1. In response to an inductive interaction mediated through Wnt/MAP kinase signaling pathways, POP-1, a Lef/Tcf-type transcription factor, restricts end-1 transcription to the posterior daughter of the mesendoderm progenitor (EMS cell), resulting in activation of endoderm differentiation by the SKN-1 and MED-1/2 transcription factors. We purified a factor from semi-synchronized early embryos that binds to an end-1 cis regulatory region critical for its endoderm-specific expression. Mass spectrometry identified this protein, PLP-1, as a C. elegans orthologue of the vertebrate pur alpha transcription factor. Expression of end-1 is attenuated in embryos depleted for PLP-1. While removal of plp-1 activity alone does not prevent endoderm development, it strongly enhances the loss of endoderm in mutants defective for the Wnt pathway. In contrast, loss of PLP-1 function does not synergize with mutants in the endoderm-inducing MAPK pathway. Moreover, nuclear localization of PLP-1 during interphase requires components of the MAPK pathway, suggesting that PLP-1 is influenced by MAPK signaling. PLP-1 is transiently asymmetrically distributed during cell divisions, with higher levels in the chromatin of the future posterior daughter of EMS and other dividing cells shortly after mitosis compared to that in their sisters. These findings imply that PLP-1 acts as a transcriptional activator of end-1 expression that may be modulated by MAPK signaling to promote endoderm development.


Assuntos
Proteínas de Caenorhabditis elegans , Endoderma/citologia , Sistema de Sinalização das MAP Quinases , Fatores de Transcrição , Proteínas Wnt/metabolismo , Animais , Células/citologia , Indução Embrionária , Endoderma/crescimento & desenvolvimento , Fatores de Transcrição GATA , Regulação da Expressão Gênica
20.
Science ; 320(5874): 365-9, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18420933

RESUMO

Mechanisms by which Wnt pathways integrate the organization of receptors, organelles, and cytoskeletal proteins to confer cell polarity and directional cell movement are incompletely understood. We show that acute responses to Wnt5a involve recruitment of actin, myosin IIB, Frizzled 3, and melanoma cell adhesion molecule into an intracellular structure in a melanoma cell line. In the presence of a chemokine gradient, this Wnt-mediated receptor-actin-myosin polarity (W-RAMP) structure accumulates asymmetrically at the cell periphery, where it triggers membrane contractility and nuclear movement in the direction of membrane retraction. The process requires endosome trafficking, is associated with multivesicular bodies, and is regulated by Wnt5a through the small guanosine triphosphatases Rab4 and RhoB. Thus, cell-autonomous mechanisms allow Wnt5a to control cell orientation, polarity, and directional movement in response to positional cues from chemokine gradients.


Assuntos
Polaridade Celular , Melanoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Actinas/metabolismo , Animais , Antígeno CD146/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Quimiocina CXCL12/metabolismo , Quimiotaxia , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Humanos , Melanoma/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Miosina não Muscular Tipo IIB/metabolismo , Transplante Heterólogo , Proteína Wnt-5a , Proteínas rab4 de Ligação ao GTP/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA