Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Environ Monit Assess ; 195(2): 265, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36600046

RESUMEN

The present study was intended for assessing the contamination level of various heavy metals in surface soil and groundwater around the automobile workshops situated at different locations in the Kollam District of Kerala state, India. The procured soil and groundwater samples were analyzed for cadmium, zinc, iron, lead, nickel, chromium, copper, manganese, and arsenic using atomic absorption spectrophotometer by following standard procedures. The contamination level of these metals was assessed using the pollution indices like enrichment factor (EF), geo-accumulation index (Igeo), contamination factor (CF), and pollution load index (PLI). The results revealed that the concentrations of all analyzed metals in the surface soils of the automobile workshops were higher than the control. On the contrary, the concentration of no heavy metal in the groundwater was either equal to or higher than the limit prescribed by WHO. However, the concentration levels of Fe, Pb, Cu, and Zn were either equal to or higher than the control values. Based on the Igeo, CF, and EF, it was found that the contamination intensity of the heavy metals in soil decreased in the following order: Fe > Pb > Cd > As > Cr > Zn > Cu > Ni > Mn. From the results of PLI, it was interpreted that the sampling sites S2, S4, and S5 were highly polluted. Non-contamination of underground water from the age-old workshops is the uniqueness of the present study against the other studies, which were completed in alluvial formations with inverse results. In the studied region, the groundwater is stored in the hard rock formations and its hydraulics remains different from alluvial aquifers.


Asunto(s)
Agua Subterránea , Metales Pesados , Contaminantes del Suelo , Suelo , Plomo , Monitoreo del Ambiente/métodos , Metales Pesados/análisis , Contaminantes del Suelo/análisis , Medición de Riesgo
2.
mBio ; 14(1): e0304222, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36622147

RESUMEN

The Toxoplasma inner membrane complex (IMC) is a unique organelle that plays critical roles in parasite motility, invasion, egress, and replication. The IMC is delineated into the apical, body, and basal regions, defined by proteins that localize to these distinct subcompartments. The IMC can be further segregated by proteins that localize specifically to the maternal IMC, the daughter bud IMC, or both. While the function of the maternal IMC has been better characterized, the precise roles of most daughter IMC components remain poorly understood. Here, we demonstrate that the daughter protein IMC29 plays an important role in parasite replication. We show that Δimc29 parasites exhibit severe replication defects, resulting in substantial growth defects and loss of virulence. Deletion analyses revealed that IMC29 localization is largely dependent on the N-terminal half of the protein containing four predicted coiled-coil domains while IMC29 function requires a short C-terminal helical region. Using proximity labeling, we identify eight novel IMC proteins enriched in daughter buds, significantly expanding the daughter IMC proteome. We additionally report four novel proteins with unique localizations to the interface between two parasites or to the outer face of the IMC, exposing new subregions of the organelle. Together, this work establishes IMC29 as an important early daughter bud component of replication and uncovers an array of new IMC proteins that provides important insights into this organelle. IMPORTANCE The inner membrane complex (IMC) is a conserved structure across the Apicomplexa phylum, which includes obligate intracellular parasites that cause toxoplasmosis, malaria, and cryptosporidiosis. The IMC is critical for the parasite to maintain its intracellular lifestyle, particularly in providing a scaffold for daughter bud formation during parasite replication. While many IMC proteins in the later stages of division have been identified, components of the early stages of division remain unknown. Here, we focus on the early daughter protein IMC29, demonstrating that it is crucial for faithful parasite replication and identifying specific regions of the protein that are important for its localization and function. We additionally use proximity labeling to reveal a suite of daughter-enriched IMC proteins, which represent promising candidates to further explore this IMC subcompartment.


Asunto(s)
Toxoplasma , Toxoplasmosis , Humanos , Toxoplasma/química , Proteoma/metabolismo , Núcleo Familiar , Proteínas Protozoarias/metabolismo , Toxoplasmosis/parasitología
3.
J Biol Chem ; 298(7): 102094, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35654137

RESUMEN

The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway delivers Fe-S clusters to nuclear and cytosolic Fe-S proteins involved in essential cellular functions. Although the delivery process is regulated by the availability of iron and oxygen, it remains unclear how CIA components orchestrate the cluster transfer under varying cellular environments. Here, we utilized a targeted proteomics assay for monitoring CIA factors and substrates to characterize the CIA machinery. We find that nucleotide-binding protein 1 (NUBP1/NBP35), cytosolic iron-sulfur assembly component 3 (CIAO3/NARFL), and CIA substrates associate with nucleotide-binding protein 2 (NUBP2/CFD1), a component of the CIA scaffold complex. NUBP2 also weakly associates with the CIA targeting complex (MMS19, CIAO1, and CIAO2B) indicating the possible existence of a higher order complex. Interactions between CIAO3 and the CIA scaffold complex are strengthened upon iron supplementation or low oxygen tension, while iron chelation and reactive oxygen species weaken CIAO3 interactions with CIA components. We further demonstrate that CIAO3 mutants defective in Fe-S cluster binding fail to integrate into the higher order complexes. However, these mutants exhibit stronger associations with CIA substrates under conditions in which the association with the CIA targeting complex is reduced suggesting that CIAO3 and CIA substrates may associate in complexes independently of the CIA targeting complex. Together, our data suggest that CIA components potentially form a metabolon whose assembly is regulated by environmental cues and requires Fe-S cluster incorporation in CIAO3. These findings provide additional evidence that the CIA pathway adapts to changes in cellular environment through complex reorganization.


Asunto(s)
Proteínas Hierro-Azufre , Hierro , Citosol/metabolismo , Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Azufre/metabolismo
4.
Commun Biol ; 5(1): 549, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35668117

RESUMEN

Over millions of years, eukaryotes evolved from unicellular to multicellular organisms with increasingly complex genomes and sophisticated gene expression networks. Consequently, chromatin regulators evolved to support this increased complexity. The ATP-dependent chromatin remodelers of the SWI/SNF family are multiprotein complexes that modulate nucleosome positioning and appear under different configurations, which perform distinct functions. While the composition, architecture, and activity of these subclasses are well understood in a limited number of fungal and animal model organisms, the lack of comprehensive information in other eukaryotic organisms precludes the identification of a reliable evolutionary model of SWI/SNF complexes. Here, we performed a systematic analysis using 36 species from animal, fungal, and plant lineages to assess the conservation of known SWI/SNF subunits across eukaryotes. We identified evolutionary relationships that allowed us to propose the composition of a hypothetical ancestral SWI/SNF complex in the last eukaryotic common ancestor. This last common ancestor appears to have undergone several rounds of lineage-specific subunit gains and losses, shaping the current conformation of the known subclasses in animals and fungi. In addition, our results unravel a plant SWI/SNF complex, reminiscent of the animal BAF subclass, which incorporates a set of plant-specific subunits of still unknown function.


Asunto(s)
Proteínas Cromosómicas no Histona , Factores de Transcripción , Animales , Cromatina , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Eucariontes/genética , Eucariontes/metabolismo , Estructuras de las Plantas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Sci Rep ; 12(1): 7133, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35504914

RESUMEN

microRNAs (miRNAs) are crucial for normal development and physiology. To identify factors that might coordinate with miRNAs to regulate gene expression, we used 2'O-methylated oligonucleotides to precipitate Caenorhabditis elegans let-7, miR-58, and miR-2 miRNAs and the associated proteins. A total of 211 proteins were identified through mass-spectrometry analysis of miRNA co-precipitates, which included previously identified interactors of key miRNA pathway components. Gene ontology analysis of the identified interactors revealed an enrichment for RNA binding proteins, suggesting that we captured proteins that may be involved in mRNA lifecycle. To determine which miRNA interactors are important for miRNA activity, we used RNAi to deplete putative miRNA co-factors in animals with compromised miRNA activity and looked for alterations of the miRNA mutant phenotypes. Depletion of 25 of 39 tested genes modified the miRNA mutant phenotypes in three sensitized backgrounds. Modulators of miRNA phenotypes ranged from RNA binding proteins RBD-1 and CEY-1 to metabolic factors such as DLST-1 and ECH-5, among others. The observed functional interactions suggest widespread coordination of these proteins with miRNAs to ultimately regulate gene expression. This study provides a foundation for future investigations aimed at deciphering the molecular mechanisms of miRNA-mediated gene regulation.


Asunto(s)
Proteínas de Caenorhabditis elegans , MicroARNs , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , MicroARNs/metabolismo , Interferencia de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
Mol Cancer Ther ; 21(6): 936-947, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35313331

RESUMEN

WNT signaling promotes pancreatic ductal adenocarcinoma (PDAC) through diverse effects on proliferation, differentiation, survival, and stemness. A subset of PDAC with inactivating mutations in ring finger protein 43 (RNF43) show growth dependency on autocrine WNT ligand signaling and are susceptible to agents that block WNT ligand acylation by Porcupine O-acyltransferase, which is required for proper WNT ligand processing and secretion. For this study, global transcriptomic, proteomic, and metabolomic analyses were performed to explore the therapeutic response of RNF43-mutant PDAC to the Porcupine inhibitor (PORCNi) LGK974. LGK974 disrupted cellular bioenergetics and mitochondrial function through actions that included rapid mitochondrial depolarization, reduced mitochondrial content, and inhibition of oxidative phosphorylation and tricarboxylic acid cycle. LGK974 also broadly altered transcriptional activity, downregulating genes involved in cell cycle, nucleotide metabolism, and ribosomal biogenesis and upregulating genes involved in epithelial-mesenchymal transition, hypoxia, endocytosis, and lysosomes. Autophagy and lysosomal activity were augmented in response to LGK974, which synergistically inhibited tumor cell viability in combination with chloroquine. Autocrine WNT ligand signaling dictates metabolic dependencies in RNF43-mutant PDAC through a combination of transcription dependent and independent effects linked to mitochondrial health and function. Metabolic adaptations to mitochondrial damage and bioenergetic stress represent potential targetable liabilities in combination with PORCNi for the treatment of WNT ligand-addicted PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Vía de Señalización Wnt , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/genética , Aciltransferasas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Proliferación Celular , Homeostasis , Humanos , Ligandos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteómica , Neoplasias Pancreáticas
7.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34494556

RESUMEN

Lipin 1 regulates cellular lipid homeostasis through roles in glycerolipid synthesis (through phosphatidic acid phosphatase activity) and transcriptional coactivation. Lipin 1-deficient individuals exhibit episodic disease symptoms that are triggered by metabolic stress, such as stress caused by prolonged fasting. We sought to identify critical lipin 1 activities during fasting. We determined that lipin 1 deficiency induces widespread alternative mRNA splicing in liver during fasting, much of which is normalized by refeeding. The role of lipin 1 in mRNA splicing was largely independent of its enzymatic function. We identified interactions between lipin 1 and spliceosome proteins, as well as a requirement for lipin 1 to maintain homeostatic levels of spliceosome small nuclear RNAs and specific RNA splicing factors. In fasted Lpin1-/- liver, we identified a correspondence between alternative splicing of phospholipid biosynthetic enzymes and dysregulated phospholipid levels; splicing patterns and phospholipid levels were partly normalized by feeding. Thus, lipin 1 influences hepatic lipid metabolism through mRNA splicing, as well as through enzymatic and transcriptional activities, and fasting exacerbates the deleterious effects of lipin 1 deficiency on metabolic homeostasis.


Asunto(s)
Adaptación Fisiológica/genética , Ayuno/fisiología , Metabolismo de los Lípidos/genética , Hígado/metabolismo , ARN Mensajero/genética , Empalme Alternativo , Animales , Células Cultivadas , Femenino , Humanos , Hígado/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Fosfatidato Fosfatasa , Empalme del ARN , Factores de Transcripción/genética
8.
J Proteome Res ; 20(9): 4318-4330, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34342229

RESUMEN

G-protein-coupled receptors (GPCRs) initiate intracellular signaling events through heterotrimeric G-protein α-subunits (Gα) and the ßγ-subunit dimer (Gßγ). In this study, we utilized mass spectrometry to identify novel regulators of Gßγ signaling in human cells. This prompted our characterization of KCTD2 and KCTD5, two related potassium channel tetramerization domain (KCTD) proteins that specifically recognize Gßγ. We demonstrated that these KCTD proteins are substrate adaptors for a multisubunit CUL3-RING ubiquitin ligase, in which a KCTD2-KCTD5 hetero-oligomer associates with CUL3 through KCTD5 subunits and recruits Gßγ through both KCTD proteins in response to G-protein activation. These KCTD proteins promote monoubiquitination of lysine-23 within Gß1/2in vitro and in HEK-293 cells. Depletion of these adaptors from cancer cell lines sharply impairs downstream signaling. Together, our studies suggest that a KCTD2-KCTD5-CUL3-RING E3 ligase recruits Gßγ in response to signaling, monoubiquitinates lysine-23 within Gß1/2, and regulates Gßγ effectors to modulate downstream signal transduction.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas , Ubiquitina-Proteína Ligasas , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Células HEK293 , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Canales de Potasio , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
9.
RNA Biol ; 18(12): 2576-2593, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34105434

RESUMEN

During spliceosome assembly, interactions that bring the 5' and 3' ends of an intron in proximity are critical for the production of mature mRNA. Here, we report synergistic roles for the stem-loops 3 (SL3) and 4 (SL4) of the human U1 small nuclear RNA (snRNA) in maintaining the optimal U1 snRNP function, and formation of cross-intron contact with the U2 snRNP. We find that SL3 and SL4 bind distinct spliceosomal proteins and combining a U1 snRNA activity assay with siRNA-mediated knockdown, we demonstrate that SL3 and SL4 act through the RNA helicase UAP56 and the U2 protein SF3A1, respectively. In vitro analysis using UV crosslinking and splicing assays indicated that SL3 likely promotes the SL4-SF3A1 interaction leading to enhancement of A complex formation and pre-mRNA splicing. Overall, these results highlight the vital role of the distinct contacts of SL3 and SL4 in bridging the pre-mRNA bound U1 and U2 snRNPs during the early steps of human spliceosome assembly.


Asunto(s)
Conformación de Ácido Nucleico , Precursores del ARN/genética , Empalme del ARN , ARN Mensajero/genética , ARN Nuclear Pequeño/genética , Secuencia de Bases , Humanos , Intrones , Precursores del ARN/química , ARN Mensajero/química , ARN Nuclear Pequeño/química
10.
J Med Chem ; 64(8): 4857-4869, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33821636

RESUMEN

LONP1 is an AAA+ protease that maintains mitochondrial homeostasis by removing damaged or misfolded proteins. Elevated activity and expression of LONP1 promotes cancer cell proliferation and resistance to apoptosis-inducing reagents. Despite the importance of LONP1 in human biology and disease, very few LONP1 inhibitors have been described in the literature. Herein, we report the development of selective boronic acid-based LONP1 inhibitors using structure-based drug design as well as the first structures of human LONP1 bound to various inhibitors. Our efforts led to several nanomolar LONP1 inhibitors with little to no activity against the 20S proteasome that serve as tool compounds to investigate LONP1 biology.


Asunto(s)
Proteasas ATP-Dependientes/antagonistas & inhibidores , Diseño de Fármacos , Proteínas Mitocondriales/antagonistas & inhibidores , Inhibidores de Proteasas/química , Proteasas ATP-Dependientes/metabolismo , Sitios de Unión , Ácidos Borónicos/química , Ácidos Borónicos/metabolismo , Ácidos Borónicos/farmacología , Bortezomib/química , Bortezomib/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Proteínas Mitocondriales/metabolismo , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad
11.
Mol Cell ; 78(5): 862-875.e8, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32348780

RESUMEN

Nuclear RNA interference (RNAi) pathways work together with histone modifications to regulate gene expression and enact an adaptive response to transposable RNA elements. In the germline, nuclear RNAi can lead to trans-generational epigenetic inheritance (TEI) of gene silencing. We identified and characterized a family of nuclear Argonaute-interacting proteins (ENRIs) that control the strength and target specificity of nuclear RNAi in C. elegans, ensuring faithful inheritance of epigenetic memories. ENRI-1/2 prevent misloading of the nuclear Argonaute NRDE-3 with small RNAs that normally effect maternal piRNAs, which prevents precocious nuclear translocation of NRDE-3 in the early embryo. Additionally, they are negative regulators of nuclear RNAi triggered from exogenous sources. Loss of ENRI-3, an unstable protein expressed mostly in the male germline, misdirects the RNAi response to transposable elements and impairs TEI. The ENRIs determine the potency and specificity of nuclear RNAi responses by gating small RNAs into specific nuclear Argonautes.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Silenciador del Gen/fisiología , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Núcleo Celular/metabolismo , Células Germinativas/metabolismo , Proteínas Nucleares/metabolismo , Interferencia de ARN/fisiología , ARN Bicatenario/metabolismo , ARN Nuclear/metabolismo , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética
12.
PLoS Biol ; 17(10): e3000475, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31584943

RESUMEN

The Toxoplasma gondii inner membrane complex (IMC) is an important organelle involved in parasite motility and replication. The IMC resides beneath the parasite's plasma membrane and is composed of both membrane and cytoskeletal components. Although the protein composition of the IMC is becoming better understood, the protein-protein associations that enable proper functioning of the organelle remain largely unknown. Determining protein interactions in the IMC cytoskeletal network is particularly challenging, as disrupting the cytoskeleton requires conditions that disrupt protein complexes. To circumvent this problem, we demonstrate the application of a photoreactive unnatural amino acid (UAA) crosslinking system to capture protein interactions in the native intracellular environment. In addition to identifying binding partners, the UAA approach maps the binding interface of the bait protein used for crosslinking, providing structural information of the interacting proteins. We apply this technology to the essential IMC protein ILP1 and demonstrate that distinct regions of its C-terminal coiled-coil domain crosslink to the alveolins IMC3 and IMC6, as well as IMC27. We also show that the IMC3 C-terminal domain and the IMC6 N-terminal domain are necessary for binding to ILP1, further mapping interactions between ILP1 and the cytoskeleton. Together, this study develops a new approach to study protein-protein interactions in Toxoplasma and provides the first insight into the architecture of the cytoskeletal network of the apicomplexan IMC.


Asunto(s)
Azidas/química , Reactivos de Enlaces Cruzados/química , Proteínas del Citoesqueleto/química , Citoesqueleto/metabolismo , Membranas Intracelulares/metabolismo , Fenilalanina/análogos & derivados , Proteínas Protozoarias/química , Toxoplasma/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/genética , Citoesqueleto/ultraestructura , Expresión Génica , Membranas Intracelulares/ultraestructura , Fenilalanina/química , Procesos Fotoquímicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Toxoplasma/ultraestructura , Rayos Ultravioleta
13.
Nat Commun ; 10(1): 3916, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477705

RESUMEN

Transcription by RNA polymerase V (Pol V) in plants is required for RNA-directed DNA methylation, leading to transcriptional gene silencing. Global chromatin association of Pol V requires components of the DDR complex DRD1, DMS3 and RDM1, but the assembly process of this complex and the underlying mechanism for Pol V recruitment remain unknown. Here we show that all DDR complex components co-localize with Pol V, and we report the cryoEM structures of two complexes associated with Pol V recruitment-DR (DMS3-RDM1) and DDR' (DMS3-RDM1-DRD1 peptide), at 3.6 Å and 3.5 Å resolution, respectively. RDM1 dimerization at the center frames the assembly of the entire complex and mediates interactions between DMS3 and DRD1 with a stoichiometry of 1 DRD1:4 DMS3:2 RDM1. DRD1 binding to the DR complex induces a drastic movement of a DMS3 coiled-coil helix bundle. We hypothesize that both complexes are functional intermediates that mediate Pol V recruitment.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN de Planta/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/ultraestructura , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/ultraestructura , Microscopía por Crioelectrón , ADN de Plantas/genética , ADN de Plantas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/ultraestructura , Regulación de la Expresión Génica de las Plantas , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Unión Proteica , Conformación Proteica , ARN de Planta/química , ARN de Planta/genética
14.
Nat Chem Biol ; 15(9): 872-881, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31406370

RESUMEN

Hundreds of cellular proteins require iron cofactors for activity, and cells express systems for their assembly and distribution. Molecular details of the cytosolic iron pool used for iron cofactors are lacking, but iron chaperones of the poly(rC)-binding protein (PCBP) family play a key role in ferrous ion distribution. Here we show that, in cells and in vitro, PCBP1 coordinates iron via conserved cysteine and glutamate residues and a molecule of noncovalently bound glutathione (GSH). Proteomics analysis of PCBP1-interacting proteins identified BolA2, which functions, in complex with Glrx3, as a cytosolic [2Fe-2S] cluster chaperone. The Fe-GSH-bound form of PCBP1 complexes with cytosolic BolA2 via a bridging Fe ligand. Biochemical analysis of PCBP1 and BolA2, in cells and in vitro, indicates that PCBP1-Fe-GSH-BolA2 serves as an intermediate complex required for the assembly of [2Fe-2S] clusters on BolA2-Glrx3, thereby linking the ferrous iron and Fe-S distribution systems in cells.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Proteínas/metabolismo , Antibacterianos/farmacología , Proteínas Portadoras , Citosol/metabolismo , Proteínas de Unión al ADN , Doxiciclina/farmacología , Compuestos Férricos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Proteínas/genética , Compuestos de Amonio Cuaternario/farmacología , Proteínas de Unión al ARN
15.
Mol Cell ; 75(2): 382-393.e5, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31229404

RESUMEN

The iron-sensing protein FBXL5 is the substrate adaptor for a SKP1-CUL1-RBX1 E3 ubiquitin ligase complex that regulates the degradation of iron regulatory proteins (IRPs). Here, we describe a mechanism of FBXL5 regulation involving its interaction with the cytosolic Fe-S cluster assembly (CIA) targeting complex composed of MMS19, FAM96B, and CIAO1. We demonstrate that the CIA-targeting complex promotes the ability of FBXL5 to degrade IRPs. In addition, the FBXL5-CIA-targeting complex interaction is regulated by oxygen (O2) tension displaying a robust association in 21% O2 that is severely diminished in 1% O2 and contributes to O2-dependent regulation of IRP degradation. Together, these data identify a novel oxygen-dependent signaling axis that links IRP-dependent iron homeostasis with the Fe-S cluster assembly machinery.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Chaperonas Moleculares/genética , Complejos Multiproteicos/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Proteínas de Ciclo Celular/química , Proteínas F-Box/química , Células HeLa , Homeostasis , Humanos , Hierro/metabolismo , Proteínas Reguladoras del Hierro/genética , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Chaperonas Moleculares/química , Complejos Multiproteicos/química , Oxígeno/metabolismo , Proteolisis , Factores de Transcripción/genética , Complejos de Ubiquitina-Proteína Ligasa/química
17.
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
18.
Sci Rep ; 8(1): 10964, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-30006618

RESUMEN

This paper has been retracted at the request of the authors.

19.
Proc Natl Acad Sci U S A ; 115(19): E4473-E4482, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686080

RESUMEN

Prostate cancer is a heterogeneous disease composed of divergent molecular and histologic subtypes, including prostate adenocarcinoma (PrAd) and neuroendocrine prostate cancer (NEPC). While PrAd is the major histology in prostate cancer, NEPC can evolve from PrAd as a mechanism of treatment resistance that involves a transition from an epithelial to a neurosecretory cancer phenotype. Cell surface markers are often associated with specific cell lineages and differentiation states in normal development and cancer. Here, we show that PrAd and NEPC can be broadly discriminated by cell-surface profiles based on the analysis of prostate cancer gene expression datasets. To overcome a dependence on predictions of human cell-surface genes and an assumed correlation between mRNA levels and protein expression, we integrated transcriptomic and cell-surface proteomic data generated from a panel of prostate cancer cell lines to nominate cell-surface markers associated with these cancer subtypes. FXYD3 and CEACAM5 were validated as cell-surface antigens enriched in PrAd and NEPC, respectively. Given the lack of effective treatments for NEPC, CEACAM5 appeared to be a promising target for cell-based immunotherapy. As a proof of concept, engineered chimeric antigen receptor T cells targeting CEACAM5 induced antigen-specific cytotoxicity in NEPC cell lines. Our findings demonstrate that the surfaceomes of PrAd and NEPC reflect unique cancer differentiation states and broadly represent vulnerabilities amenable to therapeutic targeting.


Asunto(s)
Antígenos de Superficie/análisis , Antígenos de Superficie/inmunología , Carcinoma Neuroendocrino/terapia , Neoplasias de la Próstata/terapia , Proteoma/análisis , Linfocitos T/trasplante , Transcriptoma , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Antígeno Carcinoembrionario/metabolismo , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/inmunología , Carcinoma Neuroendocrino/metabolismo , Diferenciación Celular , Células Cultivadas , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Próstata/inmunología , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Proteoma/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
20.
Mol Cell Proteomics ; 17(11): 2229-2241, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29444981

RESUMEN

The flagellated protozoan parasite Trichomonas vaginalis is the etiologic agent of trichomoniasis, the most common non-viral sexually transmitted infection worldwide. As an obligate extracellular pathogen, adherence to epithelial cells is critical for parasite survival within the human host and a better understanding of this process is a prerequisite for the development of therapies to combat infection. In this sense, recent work has shown S-acylation as a key modification that regulates pathogenesis in different protozoan parasites. However, there are no reports indicating whether this post-translational modification is a mechanism operating in T. vaginalis In order to study the extent and function of S-acylation in T. vaginalis biology, we undertook a proteomic study to profile the full scope of S-acylated proteins in this parasite and reported the identification of 363 proteins involved in a variety of biological processes such as protein transport, pathogenesis related and signaling, among others. Importantly, treatment of parasites with the palmitoylation inhibitor 2-bromopalmitate causes a significant decrease in parasite: parasite aggregation as well as adherence to host cells suggesting that palmitoylation could be modifying proteins that are key regulators of Trichomonas vaginalis pathogenesis.


Asunto(s)
Lipoilación , Proteínas Protozoarias/metabolismo , Trichomonas vaginalis/metabolismo , Adhesividad , Secuencia de Aminoácidos , Ontología de Genes , Células HeLa , Humanos , Dominios Proteicos , Proteoma/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA