Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 592(7856): 794-798, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33854239

RESUMEN

The initiation of cell division integrates a large number of intra- and extracellular inputs. D-type cyclins (hereafter, cyclin D) couple these inputs to the initiation of DNA replication1. Increased levels of cyclin D promote cell division by activating cyclin-dependent kinases 4 and 6 (hereafter, CDK4/6), which in turn phosphorylate and inactivate the retinoblastoma tumour suppressor. Accordingly, increased levels and activity of cyclin D-CDK4/6 complexes are strongly linked to unchecked cell proliferation and cancer2,3. However, the mechanisms that regulate levels of cyclin D are incompletely understood4,5. Here we show that autophagy and beclin 1 regulator 1 (AMBRA1) is the main regulator of the degradation of cyclin D. We identified AMBRA1 in a genome-wide screen to investigate the genetic basis of  the response to CDK4/6 inhibition. Loss of AMBRA1 results in high levels of cyclin D in cells and in mice, which promotes proliferation and decreases sensitivity to CDK4/6 inhibition. Mechanistically, AMBRA1 mediates ubiquitylation and proteasomal degradation of cyclin D as a substrate receptor for the cullin 4 E3 ligase complex. Loss of AMBRA1 enhances the growth of lung adenocarcinoma in a mouse model, and low levels of AMBRA1 correlate with worse survival in patients with lung adenocarcinoma. Thus, AMBRA1 regulates cellular levels of cyclin D, and contributes to cancer development and the response of cancer cells to CDK4/6 inhibitors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ciclina D/metabolismo , Adenocarcinoma del Pulmón/genética , Animales , División Celular , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Genes Supresores de Tumor , Humanos , Neoplasias Pulmonares/genética , Ratones , Piperazinas/farmacología , Piridinas/farmacología , Células U937 , Ubiquitinación
2.
Development ; 149(11)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35695185

RESUMEN

In the developing subpallium, the fate decision between neurons and glia is driven by expression of Dlx1/2 or Olig1/2, respectively, two sets of transcription factors with a mutually repressive relationship. The mechanism by which Dlx1/2 repress progenitor and oligodendrocyte fate, while promoting transcription of genes needed for differentiation, is not fully understood. We identified a motif within DLX1 that binds RBBP4, a NuRD complex subunit. ChIP-seq studies of genomic occupancy of DLX1 and six different members of the NuRD complex show that DLX1 and NuRD colocalize to putative regulatory elements enriched near other transcription factor genes. Loss of Dlx1/2 leads to dysregulation of genome accessibility at putative regulatory elements near genes repressed by Dlx1/2, including Olig2. Consequently, heterozygosity of Dlx1/2 and Rbbp4 leads to an increase in the production of OLIG2+ cells. These findings highlight the importance of the interplay between transcription factors and chromatin remodelers in regulating cell-fate decisions.


Asunto(s)
Proteínas de Homeodominio , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Diferenciación Celular/genética , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Mol Cell Proteomics ; 22(5): 100541, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37019383

RESUMEN

Apolipoprotein (apo) E4 is the major genetic risk factor for Alzheimer's disease. While neurons generally produce a minority of the apoE in the central nervous system, neuronal expression of apoE increases dramatically in response to stress and is sufficient to drive pathology. Currently, the molecular mechanisms of how apoE4 expression may regulate pathology are not fully understood. Here, we expand upon our previous studies measuring the impact of apoE4 on protein abundance to include the analysis of protein phosphorylation and ubiquitylation signaling in isogenic Neuro-2a cells expressing apoE3 or apoE4. ApoE4 expression resulted in a dramatic increase in vasodilator-stimulated phosphoprotein (VASP) S235 phosphorylation in a protein kinase A (PKA)-dependent manner. This phosphorylation disrupted VASP interactions with numerous actin cytoskeletal and microtubular proteins. Reduction of VASP S235 phosphorylation via PKA inhibition resulted in a significant increase in filopodia formation and neurite outgrowth in apoE4-expressing cells, exceeding levels observed in apoE3-expressing cells. Our results highlight the pronounced and diverse impact of apoE4 on multiple modes of protein regulation and identify protein targets to restore apoE4-related cytoskeletal defects.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Actinas/metabolismo , Enfermedad de Alzheimer/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Fosforilación , Proteómica , Animales , Ratones
4.
J Biol Chem ; 297(1): 100907, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34166681

RESUMEN

Endosomal signaling downstream of G-protein-coupled receptors (GPCRs) has emerged as a novel paradigm with important pharmacological and physiological implications. However, our knowledge of the functional consequences of intracellular signaling is incomplete. To begin to address this gap, we combined an optogenetic approach for site-specific generation of the prototypical second messenger generated by active GPCRs, cyclic AMP (cAMP), with unbiased mass-spectrometry-based analysis of the phosphoproteome. We identified 218 unique, high-confidence sites whose phosphorylation is either increased or decreased in response to cAMP elevation. We next determined that the same amount of cAMP produced from the endosomal membrane led to more robust changes in phosphorylation than the plasma membrane. Remarkably, this was true for the entire repertoire of 218 identified targets and irrespective of their annotated subcellular localizations (endosome, cell surface, nucleus, cytosol). Furthermore, we identified a particularly strong endosome bias for a subset of proteins that are dephosphorylated in response to cAMP. Through bioinformatics analysis, we established these targets as putative substrates for protein phosphatase 2A (PP2A), and we propose compartmentalized activation of PP2A by cAMP-responsive kinases as the likely underlying mechanism. Altogether, our study extends the concept that endosomal signaling is a significant functional contributor to cellular responsiveness to cAMP by establishing a unique role for localized cAMP production in defining categorically distinct phosphoresponses.


Asunto(s)
AMP Cíclico/metabolismo , Endosomas/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Animales , Células HEK293 , Humanos , Fosfoproteínas/química , Fosforilación , Dominios Proteicos , Proteína Fosfatasa 2/metabolismo , Proteoma/química
5.
EMBO J ; 37(18)2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30166453

RESUMEN

Multi-subunit cullin-RING ligases (CRLs) are the largest family of ubiquitin E3 ligases in humans. CRL activity is tightly regulated to prevent unintended substrate degradation or autocatalytic degradation of CRL subunits. Using a proteomics strategy, we discovered that CRL4AMBRA1 (CRL substrate receptor denoted in superscript) targets Elongin C (ELOC), the essential adapter protein of CRL5 complexes, for polyubiquitination and degradation. We showed that the ubiquitin ligase function of CRL4AMBRA1 is required to disrupt the assembly and attenuate the ligase activity of human CRL5SOCS3 and HIV-1 CRL5VIF complexes as AMBRA1 depletion leads to hyperactivation of both CRL5 complexes. Moreover, CRL4AMBRA1 modulates interleukin-6/STAT3 signaling and HIV-1 infectivity that are regulated by CRL5SOCS3 and CRL5VIF, respectively. Thus, by discovering a substrate of CRL4AMBRA1, ELOC, the shared adapter of CRL5 ubiquitin ligases, we uncovered a novel CRL cross-regulation pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Elonguina/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Proteolisis , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Elonguina/genética , Células HEK293 , Infecciones por VIH/genética , VIH-1/genética , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
6.
Proc Natl Acad Sci U S A ; 112(45): 13976-81, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26504226

RESUMEN

Although a variety of genetic alterations have been found across cancer types, the identification and functional characterization of candidate driver genetic lesions in an individual patient and their translation into clinically actionable strategies remain major hurdles. Here, we use whole genome sequencing of a prostate cancer tumor, computational analyses, and experimental validation to identify and predict novel oncogenic activity arising from a point mutation in the phosphatase and tensin homolog (PTEN) tumor suppressor protein. We demonstrate that this mutation (p.A126G) produces an enzymatic gain-of-function in PTEN, shifting its function from a phosphoinositide (PI) 3-phosphatase to a phosphoinositide (PI) 5-phosphatase. Using cellular assays, we demonstrate that this gain-of-function activity shifts cellular phosphoinositide levels, hyperactivates the PI3K/Akt cell proliferation pathway, and exhibits increased cell migration beyond canonical PTEN loss-of-function mutants. These findings suggest that mutationally modified PTEN can actively contribute to well-defined hallmarks of cancer. Lastly, we demonstrate that these effects can be substantially mitigated through chemical PI3K inhibitors. These results demonstrate a new dysfunction paradigm for PTEN cancer biology and suggest a potential framework for the translation of genomic data into actionable clinical strategies for targeted patient therapy.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Neoplasias/genética , Fosfohidrolasa PTEN/genética , Monoéster Fosfórico Hidrolasas/genética , Neoplasias de la Próstata/genética , Análisis de Varianza , Animales , Secuencia de Bases , Células CHO , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Biología Computacional/métodos , Cricetinae , Cricetulus , Humanos , Immunoblotting , Masculino , Microscopía Fluorescente , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Técnicas de Placa-Clamp , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Análisis de Secuencia de ADN
7.
Mol Pharmacol ; 91(2): 145-156, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27879340

RESUMEN

The ability of chemically distinct ligands to produce different effects on the same G protein-coupled receptor (GPCR) has interesting therapeutic implications, but, if excessively propagated downstream, would introduce biologic noise compromising cognate ligand detection. We asked whether cells have the ability to limit the degree to which chemical diversity imposed at the ligand-GPCR interface is propagated to the downstream signal. We carried out an unbiased analysis of the integrated cellular response elicited by two chemically and pharmacodynamically diverse ß-adrenoceptor agonists, isoproterenol and salmeterol. We show that both ligands generate an identical integrated response, and that this stereotyped output requires endocytosis. We further demonstrate that the endosomal ß2-adrenergic receptor signal confers uniformity on the downstream response because it is highly sensitive and saturable. Based on these findings, we propose that GPCR signaling from endosomes functions as a biologic noise filter to enhance reliability of cognate ligand detection.


Asunto(s)
Endocitosis , Receptores Acoplados a Proteínas G/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Células HEK293 , Humanos , Isoproterenol/farmacología , Ligandos , Espectrometría de Masas , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteoma/metabolismo , Proteómica , Receptores Adrenérgicos beta 2/metabolismo , Xinafoato de Salmeterol/farmacología , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
Mol Cell Proteomics ; 14(1): 162-76, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25381059

RESUMEN

Although histone acetylation and deacetylation machineries (HATs and HDACs) regulate important aspects of cell function by targeting histone tails, recent work highlights that non-histone protein acetylation is also pervasive in eukaryotes. Here, we use quantitative mass-spectrometry to define acetylations targeted by the sirtuin family, previously implicated in the regulation of non-histone protein acetylation. To identify HATs that promote acetylation of these sites, we also performed this analysis in gcn5 (SAGA) and esa1 (NuA4) mutants. We observed strong sequence specificity for the sirtuins and for each of these HATs. Although the Gcn5 and Esa1 consensus sequences are entirely distinct, the sirtuin consensus overlaps almost entirely with that of Gcn5, suggesting a strong coordination between these two regulatory enzymes. Furthermore, by examining global acetylation in an ada2 mutant, which dissociates Gcn5 from the SAGA complex, we found that a subset of Gcn5 targets did not depend on an intact SAGA complex for targeting. Our work provides a framework for understanding how HAT and HDAC enzymes collaborate to regulate critical cellular processes related to growth and division.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Sirtuinas/metabolismo , Acetilación , Histona Desacetilasas/metabolismo , Proteoma
9.
Biochemistry ; 54(29): 4423-6, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26161950

RESUMEN

The SCF ubiquitin ligase associates with substrates through its F-box protein adaptor. Substrates are typically recognized through a defined phosphodegron. Here, we characterize the interaction of the F-box protein Saf1 with Prb1, one of its vacuolar protease substrates. We show that Saf1 binds the mature protein but ubiquitinates only the zymogen precursor. The ubiquitinated lysine was found to be in a peptide eliminated from the mature protein. Mutations that eliminate the catalytic activity of Prb1, or the related substrate Prc1, block Saf1 targeting of the zymogen precursor. Our data suggest that Saf1 does not require a conventional degron as do other F-box proteins but instead recognizes the catalytic site itself.


Asunto(s)
Endopeptidasas/química , Proteínas F-Box/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Endopeptidasas/fisiología , Unión Proteica , Proteolisis , Proteínas de Saccharomyces cerevisiae/fisiología
10.
Nat Commun ; 14(1): 6030, 2023 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-37758692

RESUMEN

Influenza A Virus (IAV) is a recurring respiratory virus with limited availability of antiviral therapies. Understanding host proteins essential for IAV infection can identify targets for alternative host-directed therapies (HDTs). Using affinity purification-mass spectrometry and global phosphoproteomic and protein abundance analyses using three IAV strains (pH1N1, H3N2, H5N1) in three human cell types (A549, NHBE, THP-1), we map 332 IAV-human protein-protein interactions and identify 13 IAV-modulated kinases. Whole exome sequencing of patients who experienced severe influenza reveals several genes, including scaffold protein AHNAK, with predicted loss-of-function variants that are also identified in our proteomic analyses. Of our identified host factors, 54 significantly alter IAV infection upon siRNA knockdown, and two factors, AHNAK and coatomer subunit COPB1, are also essential for productive infection by SARS-CoV-2. Finally, 16 compounds targeting our identified host factors suppress IAV replication, with two targeting CDK2 and FLT3 showing pan-antiviral activity across influenza and coronavirus families. This study provides a comprehensive network model of IAV infection in human cells, identifying functional host targets for pan-viral HDT.


Asunto(s)
COVID-19 , Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Humana , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Proteómica , Replicación Viral/genética , SARS-CoV-2 , Antivirales/metabolismo , Interacciones Huésped-Patógeno/genética
11.
Cell Rep ; 39(2): 110690, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417684

RESUMEN

Viruses must effectively remodel host cellular pathways to replicate and evade immune defenses, and they must do so with limited genomic coding capacity. Targeting post-translational modification (PTM) pathways provides a mechanism by which viruses can broadly and rapidly transform a hostile host environment into a hospitable one. We use mass spectrometry-based proteomics to quantify changes in protein abundance and two PTM types-phosphorylation and ubiquitination-in response to HIV-1 infection with viruses harboring targeted deletions of a subset of HIV-1 genes. PTM analysis reveals a requirement for Aurora kinase activity in HIV-1 infection and identified putative substrates of a phosphatase that is degraded during infection. Finally, we demonstrate that the HIV-1 Vpr protein inhibits histone H1 ubiquitination, leading to defects in DNA repair.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , VIH-1/genética , Humanos , Procesamiento Proteico-Postraduccional , Proteómica , Ubiquitinación
12.
J Proteome Res ; 10(10): 4692-702, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21815628

RESUMEN

The increase in adipose tissue mass arises in part from progressive lipid loading and triglyceride accumulation in adipocytes. Enlarged adipocytes produce the highest levels of pro-inflammatory molecules and reactive oxygen species (ROS). Since mitochondria are the site for major metabolic processes (e.g., TCA cycle) that govern the extent of triglyceride accumulation as well as the primary site of ROS generation, we quantitatively investigated changes in the adipocyte mitochondrial proteome during different stages of differentiation and enlargement. Mitochondrial proteins from 3T3-L1 adipocytes at different stages of lipid accumulation (days 0-18) were digested and labeled using the iTRAQ 8-plex kit. The labeled peptides were fractionated using a liquid phase isoelectric fractionation system (MSWIFT) to increase the depth of proteome coverage and analyzed using LC-MS/MS. A total of 631 proteins in the mitochondrial fraction, including endoplasmic reticulum-associated and golgi-related mitochondrial proteins, were identified and classified into 12 functional categories. A total of 123 proteins demonstrated a statistically significant change in expression in at least one of the time points over the course of the experiment. The identified proteins included enzymes and transporters involved in the TCA cycle, fatty acid oxidation, and ATP synthesis. Our results indicate that cultured adipocytes enter a state of metabolic-overdrive where increased flux through the TCA cycle and increased fatty acid oxidation occur simultaneously. The proteomic data also suggest that accumulation of reduced electron carriers and the resultant oxidative stress may be attractive targets for modulating adipocyte function in metabolic disorders.


Asunto(s)
Adipocitos/citología , Mitocondrias/metabolismo , Proteómica/métodos , Células 3T3-L1 , Adenosina Trifosfato/metabolismo , Adipocitos/metabolismo , Animales , Diferenciación Celular , Aumento de la Célula , Ciclo del Ácido Cítrico , Ácidos Grasos/metabolismo , Ratones , Modelos Biológicos , Estrés Oxidativo , Especies Reactivas de Oxígeno
13.
J Alzheimers Dis ; 68(3): 991-1011, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30883359

RESUMEN

Apolipoprotein (apo) E4, the major genetic risk factor for Alzheimer's disease (AD), alters mitochondrial function and metabolism early in AD pathogenesis. When injured or stressed, neurons increase apoE synthesis. Because of its structural difference from apoE3, apoE4 undergoes neuron-specific proteolysis, generating fragments that enter the cytosol, interact with mitochondria, and cause neurotoxicity. However, apoE4's effect on mitochondrial respiration and metabolism is not understood in detail. Here we used biochemical assays and proteomic profiling to more completely characterize the effects of apoE4 on mitochondrial function and cellular metabolism in Neuro-2a neuronal cells stably expressing apoE4 or apoE3. Under basal conditions, apoE4 impaired respiration and increased glycolysis, but when challenged or stressed, apoE4-expressing neurons had 50% less reserve capacity to generate ATP to meet energy requirements than apoE3-expressing neurons. ApoE4 expression also decreased the NAD+/NADH ratio and increased the levels of reactive oxygen species and mitochondrial calcium. Global proteomic profiling revealed widespread changes in mitochondrial processes in apoE4 cells, including reduced levels of numerous respiratory complex subunits and major disruptions to all detected subunits in complex V (ATP synthase). Also altered in apoE4 cells were levels of proteins related to mitochondrial endoplasmic reticulum-associated membranes, mitochondrial fusion/fission, mitochondrial protein translocation, proteases, and mitochondrial ribosomal proteins. ApoE4-induced bioenergetic deficits led to extensive metabolic rewiring, but despite numerous cellular adaptations, apoE4-expressing neurons remained vulnerable to metabolic stress. Our results provide insights into potential molecular targets of therapies to correct apoE4-associated mitochondrial dysfunction and altered cellular metabolism.


Asunto(s)
Apolipoproteína E4/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Proteoma/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Tumoral , Metabolismo Energético , Ratones , NAD/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico , Transcriptoma
14.
Nat Microbiol ; 4(6): 985-995, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30833725

RESUMEN

West Nile virus (WNV) is an emerging mosquito-borne flavivirus, related to dengue virus and Zika virus. To gain insight into host pathways involved in WNV infection, we performed a systematic affinity-tag purification mass spectrometry (APMS) study to identify 259 WNV-interacting human proteins. RNA interference screening revealed 26 genes that both interact with WNV proteins and influence WNV infection. We found that WNV, dengue and Zika virus capsids interact with a conserved subset of proteins that impact infection. These include the exon-junction complex (EJC) recycling factor PYM1, which is antiviral against all three viruses. The EJC has roles in nonsense-mediated decay (NMD), and we found that both the EJC and NMD are antiviral and the EJC protein RBM8A directly binds WNV RNA. To counteract this, flavivirus infection inhibits NMD and the capsid-PYM1 interaction interferes with EJC protein function and localization. Depletion of PYM1 attenuates RBM8A binding to viral RNA, suggesting that WNV sequesters PYM1 to protect viral RNA from decay. Together, these data suggest a complex interplay between the virus and host in regulating NMD and the EJC.


Asunto(s)
Antivirales/farmacología , Infecciones por Flavivirus/tratamiento farmacológico , Proteínas Virales/genética , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/genética , Proteínas de la Cápside , Proteínas Portadoras , Codón sin Sentido , Virus del Dengue/genética , Exones , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Mapas de Interacción de Proteínas , Interferencia de ARN , ARN Viral , Proteínas de Unión al ARN , Proteínas Virales/fisiología , Virus del Nilo Occidental/patogenicidad , Virus Zika/genética
15.
Cell Rep ; 28(9): 2317-2330.e8, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31461649

RESUMEN

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor and bi-functional lipid and protein phosphatase. We report that the metabolic regulator pyruvate dehydrogenase kinase1 (PDHK1) is a synthetic-essential gene in PTEN-deficient cancer and normal cells. The PTEN protein phosphatase dephosphorylates nuclear factor κB (NF-κB)-activating protein (NKAP) and limits NFκB activation to suppress expression of PDHK1, a NF-κB target gene. Loss of the PTEN protein phosphatase upregulates PDHK1 to induce aerobic glycolysis and PDHK1 cellular dependence. PTEN-deficient human tumors harbor increased PDHK1, a biomarker of decreased patient survival. This study uncovers a PTEN-regulated signaling pathway and reveals PDHK1 as a potential target in PTEN-deficient cancers.


Asunto(s)
Neoplasias/metabolismo , Fosfohidrolasa PTEN/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Animales , Línea Celular Tumoral , Femenino , Glucólisis , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/metabolismo , Neoplasias/genética , Neoplasias/patología , Fosfohidrolasa PTEN/economía , Fosfohidrolasa PTEN/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Proteínas Represoras/metabolismo
16.
G3 (Bethesda) ; 8(12): 3931-3944, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30377154

RESUMEN

The highly conserved DNA damage response (DDR) pathway monitors the genomic integrity of the cell and protects against genotoxic stresses. The apical kinases, Mec1 and Tel1 (ATR and ATM in human, respectively), initiate the DNA damage signaling cascade through the effector kinases, Rad53 and Chk1, to regulate a variety of cellular processes including cell cycle progression, DNA damage repair, chromatin remodeling, and transcription. The DDR also regulates other cellular pathways, but direct substrates and mechanisms are still lacking. Using a mass spectrometry-based phosphoproteomic screen in Saccharomyces cerevisiae, we identified novel targets of Rad53, many of which are proteins that are involved in RNA metabolism. Of the 33 novel substrates identified, we verified that 12 are directly phosphorylated by Rad53 in vitro: Xrn1, Gcd11, Rps7b, Ded1, Cho2, Pus1, Hst1, Srv2, Set3, Snu23, Alb1, and Scp160. We further characterized Xrn1, a highly conserved 5' exoribonuclease that functions in RNA degradation and the most enriched in our phosphoproteomics screen. Phosphorylation of Xrn1 by Rad53 does not appear to affect Xrn1's intrinsic nuclease activity in vitro, but may affect its activity or specificity in vivo.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2/metabolismo , Estabilidad del ARN/fisiología , ARN de Hongos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , Daño del ADN/fisiología , Reparación del ADN/fisiología , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Fosforilación/fisiología , ARN de Hongos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Especificidad por Sustrato/fisiología
17.
Elife ; 72018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29845934

RESUMEN

The HIV-1 Tat protein hijacks P-TEFb kinase to activate paused RNA polymerase II (RNAP II) at the viral promoter. Tat binds additional host factors, but it is unclear how they regulate RNAP II elongation. Here, we identify the cytoplasmic ubiquitin ligase UBE2O as critical for Tat transcriptional activity. Tat hijacks UBE2O to ubiquitinate the P-TEFb kinase inhibitor HEXIM1 of the 7SK snRNP, a fraction of which also resides in the cytoplasm bound to P-TEFb. HEXIM1 ubiquitination sequesters it in the cytoplasm and releases P-TEFb from the inhibitory 7SK complex. Free P-TEFb then becomes enriched in chromatin, a process that is also stimulated by treating cells with a CDK9 inhibitor. Finally, we demonstrate that UBE2O is critical for P-TEFb recruitment to the HIV-1 promoter. Together, the data support a unique model of elongation control where non-degradative ubiquitination of nuclear and cytoplasmic 7SK snRNP pools increases P-TEFb levels for transcriptional activation.


Asunto(s)
VIH-1/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Activación Transcripcional/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Secuencia de Aminoácidos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Cromatina/metabolismo , Diclororribofuranosil Benzoimidazol/farmacología , Células HEK293 , Células HeLa , Humanos , Modelos Biológicos , Señales de Localización Nuclear/química , Señales de Localización Nuclear/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción , Activación Transcripcional/efectos de los fármacos , Enzimas Ubiquitina-Conjugadoras/química , Ubiquitinación/efectos de los fármacos
18.
Elife ; 4: e07314, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25875391

RESUMEN

The general translation initiation factor eIF2 is a major translational control point. Multiple signaling pathways in the integrated stress response phosphorylate eIF2 serine-51, inhibiting nucleotide exchange by eIF2B. ISRIB, a potent drug-like small molecule, renders cells insensitive to eIF2α phosphorylation and enhances cognitive function in rodents by blocking long-term depression. ISRIB was identified in a phenotypic cell-based screen, and its mechanism of action remained unknown. We now report that ISRIB is an activator of eIF2B. Our reporter-based shRNA screen revealed an eIF2B requirement for ISRIB activity. Our results define ISRIB as a symmetric molecule, show ISRIB-mediated stabilization of activated eIF2B dimers, and suggest that eIF2B4 (δ-subunit) contributes to the ISRIB binding site. We also developed new ISRIB analogs, improving its EC50 to 600 pM in cell culture. By modulating eIF2B function, ISRIB promises to be an invaluable tool in proof-of-principle studies aiming to ameliorate cognitive defects resulting from neurodegenerative diseases.


Asunto(s)
Acetamidas/química , Ciclohexilaminas/química , Factor 2B Eucariótico de Iniciación/genética , Fármacos Neuroprotectores/química , Nootrópicos/química , Subunidades de Proteína/genética , Acetamidas/síntesis química , Acetamidas/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Ciclohexilaminas/síntesis química , Ciclohexilaminas/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2B Eucariótico de Iniciación/antagonistas & inhibidores , Factor 2B Eucariótico de Iniciación/metabolismo , Expresión Génica , Genes Reporteros , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Células K562 , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacología , Nootrópicos/síntesis química , Nootrópicos/farmacología , Fosforilación , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Relación Estructura-Actividad , Tapsigargina/antagonistas & inhibidores , Tapsigargina/farmacología
19.
Cell Rep ; 11(8): 1236-50, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25981045

RESUMEN

HIV-1 encodes the accessory protein Vif, which hijacks a host Cullin-RING ubiquitin ligase (CRL) complex as well as the non-canonical cofactor CBFß, to antagonize APOBEC3 antiviral proteins. Non-canonical cofactor recruitment to CRL complexes by viral factors, to date, has only been attributed to HIV-1 Vif. To further study this phenomenon, we employed a comparative approach combining proteomic, biochemical, structural, and virological techniques to investigate Vif complexes across the lentivirus genus, including primate (HIV-1 and simian immunodeficiency virus macaque [SIVmac]) and non-primate (FIV, BIV, and MVV) viruses. We find that CBFß is completely dispensable for the activity of non-primate lentiviral Vif proteins. Furthermore, we find that BIV Vif requires no cofactor and that MVV Vif requires a novel cofactor, cyclophilin A (CYPA), for stable CRL complex formation and anti-APOBEC3 activity. We propose modular conservation of Vif complexes allows for potential exaptation of functions through the acquisition of non-CRL-associated host cofactors while preserving anti-APOBEC3 activity.


Asunto(s)
Citosina Desaminasa/antagonistas & inhibidores , Productos del Gen vif/inmunología , VIH-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Desaminasas APOBEC , Animales , Citidina Desaminasa , Humanos , Unión Proteica , Ovinos , Ubiquitina-Proteína Ligasas/genética
20.
Methods Mol Biol ; 909: 259-77, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22903721

RESUMEN

Understanding changes in the expression of specific proteins and/or alterations in their posttranslational modifications is crucial to elucidating the molecular mechanisms underlying disease states such as alcoholic liver disease. Protein separation and analysis techniques such as two-dimensional electrophoresis and mass spectrometry can be used for identifying biomarker proteins that are altered during progression of alcoholic liver disease. In this chapter, we outline methods for resolving liver tissue proteins from a rodent model of alcoholic liver disease using two-dimensional electrophoresis and identifying differentially expressed proteins using mass spectrometry. In addition, since oxidative stress strongly correlates with alcoholic liver disease, we also describe methods for identifying oxidatively modified proteins from liver tissue. We specifically focus on identifying proteins that are carbonylated as protein carbonylation is a permanent modification and considered deleterious to cells. The combination of two-dimensional electrophoresis for protein resolution, mass spectrometry for protein identification, and affinity-based methods for enriching and identifying carbonylated proteins is a powerful methodology for protein biomarker identification.


Asunto(s)
Hígado Graso Alcohólico/metabolismo , Hígado/metabolismo , Proteoma/aislamiento & purificación , Animales , Biotinilación , Cromatografía de Afinidad , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Electroforesis en Gel de Poliacrilamida , Focalización Isoeléctrica , Hígado/química , Carbonilación Proteica , Proteolisis , Proteoma/química , Proteoma/metabolismo , Proteómica , Ratas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Coloración y Etiquetado , Espectrometría de Masas en Tándem , Extractos de Tejidos/química , Extractos de Tejidos/aislamiento & purificación , Tripsina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA