Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 180(2): 373-386.e15, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31955847

RESUMEN

Molecular interactions at the cellular interface mediate organized assembly of single cells into tissues and, thus, govern the development and physiology of multicellular organisms. Here, we developed a cell-type-specific, spatiotemporally resolved approach to profile cell-surface proteomes in intact tissues. Quantitative profiling of cell-surface proteomes of Drosophila olfactory projection neurons (PNs) in pupae and adults revealed global downregulation of wiring molecules and upregulation of synaptic molecules in the transition from developing to mature PNs. A proteome-instructed in vivo screen identified 20 cell-surface molecules regulating neural circuit assembly, many of which belong to evolutionarily conserved protein families not previously linked to neural development. Genetic analysis further revealed that the lipoprotein receptor LRP1 cell-autonomously controls PN dendrite targeting, contributing to the formation of a precise olfactory map. These findings highlight the power of temporally resolved in situ cell-surface proteomic profiling in discovering regulators of brain wiring.


Asunto(s)
Vías Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Proteómica/métodos , Animales , Axones/metabolismo , Encéfalo/metabolismo , Dendritas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Neurogénesis/fisiología , Nervio Olfatorio/metabolismo , Vías Olfatorias/citología , Vías Olfatorias/fisiología , Receptores de Lipoproteína/metabolismo , Olfato/fisiología
2.
Cell ; 166(5): 1295-1307.e21, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565350

RESUMEN

Cellular compartments that cannot be biochemically isolated are challenging to characterize. Here we demonstrate the proteomic characterization of the synaptic clefts that exist at both excitatory and inhibitory synapses. Normal brain function relies on the careful balance of these opposing neural connections, and understanding how this balance is achieved relies on knowledge of their protein compositions. Using a spatially restricted enzymatic tagging strategy, we mapped the proteomes of two of the most common excitatory and inhibitory synaptic clefts in living neurons. These proteomes reveal dozens of synaptic candidates and assign numerous known synaptic proteins to a specific cleft type. The molecular differentiation of each cleft allowed us to identify Mdga2 as a potential specificity factor influencing Neuroligin-2's recruitment of presynaptic neurotransmitters at inhibitory synapses.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Neuronas GABAérgicas/metabolismo , Inmunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteoma/metabolismo , Membranas Sinápticas/metabolismo , Animales , Antígenos CD/metabolismo , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Ratones , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Peroxidasa/genética , Peroxidasa/metabolismo , Proteómica , Ratas , Receptores de GABA/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tálamo/metabolismo
3.
Genes Dev ; 32(13-14): 929-943, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950492

RESUMEN

While a mutation in C9ORF72 is the most common genetic contributor to amyotrophic lateral sclerosis (ALS), much remains to be learned concerning the function of the protein normally encoded at this locus. To elaborate further on functions for C9ORF72, we used quantitative mass spectrometry-based proteomics to identify interacting proteins in motor neurons and found that its long isoform complexes with and stabilizes SMCR8, which further enables interaction with WDR41. To study the organismal and cellular functions for this tripartite complex, we generated Smcr8 loss-of-function mutant mice and found that they developed phenotypes also observed in C9orf72 loss-of-function animals, including autoimmunity. Along with a loss of tolerance for many nervous system autoantigens, we found increased lysosomal exocytosis in Smcr8 mutant macrophages. In addition to elevated surface Lamp1 (lysosome-associated membrane protein 1) expression, we also observed enhanced secretion of lysosomal components-phenotypes that we subsequently observed in C9orf72 loss-of-function macrophages. Overall, our findings demonstrate that C9ORF72 and SMCR8 have interdependent functions in suppressing autoimmunity as well as negatively regulating lysosomal exocytosis-processes of potential importance to ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Autoinmunidad/genética , Proteínas Portadoras/metabolismo , Exocitosis/genética , Lisosomas/metabolismo , Animales , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Proteínas Portadoras/genética , Regulación de la Expresión Génica/genética , Humanos , Ganglios Linfáticos/patología , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Mutación , Isoformas de Proteínas , Estabilidad Proteica , Esplenomegalia/genética
4.
EMBO Rep ; 24(12): e56997, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37975164

RESUMEN

Planar cell polarity (PCP) signaling polarizes epithelial cells within the plane of an epithelium. Core PCP signaling components adopt asymmetric subcellular localizations within cells to both polarize and coordinate polarity between cells. Achieving subcellular asymmetry requires additional effectors, including some mediating post-translational modifications of core components. Identification of such proteins is challenging due to pleiotropy. We used mass spectrometry-based proximity labeling proteomics to identify such regulators in the Drosophila wing. We identified the catalytic subunit of protein phosphatase1, Pp1-87B, and show that it regulates core protein polarization. Pp1-87B interacts with the core protein Van Gogh and at least one serine/threonine kinase, Dco/CKIε, that is known to regulate PCP. Pp1-87B modulates Van Gogh subcellular localization and directs its dephosphorylation in vivo. PNUTS, a Pp1 regulatory subunit, also modulates PCP. While the direct substrate(s) of Pp1-87B in control of PCP is not known, our data support the model that cycling between phosphorylated and unphosphorylated forms of one or more core PCP components may regulate acquisition of asymmetry. Finally, our screen serves as a resource for identifying additional regulators of PCP signaling.


Asunto(s)
Proteínas de Drosophila , Proteínas de la Membrana , Animales , Polaridad Celular/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
5.
Blood ; 137(16): 2209-2220, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33512474

RESUMEN

Casitas B-lineage lymphoma (CBL) encodes an E3 ubiquitin ligase and signaling adaptor that regulates receptor and nonreceptor tyrosine kinases. Recurrent CBL mutations occur in myeloid neoplasms, including 10% to 20% of chronic myelomonocytic leukemia (CMML) cases, and selectively disrupt the protein's E3 ubiquitin ligase activity. CBL mutations have been associated with poor prognosis, but the oncogenic mechanisms and therapeutic implications of CBL mutations remain incompletely understood. We combined functional assays and global mass spectrometry to define the phosphoproteome, CBL interactome, and mechanism of signaling activation in a panel of cell lines expressing an allelic series of CBL mutations. Our analyses revealed that increased LYN activation and interaction with mutant CBL are key drivers of enhanced CBL phosphorylation, phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1) recruitment, and downstream phosphatidylinositol 3-kinase (PI3K)/AKT signaling in CBL-mutant cells. Signaling adaptor domains of CBL, including the tyrosine kinase-binding domain, proline-rich region, and C-terminal phosphotyrosine sites, were all required for the oncogenic function of CBL mutants. Genetic ablation or dasatinib-mediated inhibition of LYN reduced CBL phosphorylation, CBL-PIK3R1 interaction, and PI3K/AKT signaling. Furthermore, we demonstrated in vitro and in vivo antiproliferative efficacy of dasatinib in CBL-mutant cell lines and primary CMML. Overall, these mechanistic insights into the molecular function of CBL mutations provide rationale to explore the therapeutic potential of LYN inhibition in CBL-mutant myeloid malignancies.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-cbl/genética , Familia-src Quinasas/metabolismo , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Humanos , Mutación , Mapas de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 117(22): 12143-12154, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32424107

RESUMEN

Proximity labeling catalyzed by promiscuous enzymes, such as TurboID, have enabled the proteomic analysis of subcellular regions difficult or impossible to access by conventional fractionation-based approaches. Yet some cellular regions, such as organelle contact sites, remain out of reach for current PL methods. To address this limitation, we split the enzyme TurboID into two inactive fragments that recombine when driven together by a protein-protein interaction or membrane-membrane apposition. At endoplasmic reticulum-mitochondria contact sites, reconstituted TurboID catalyzed spatially restricted biotinylation, enabling the enrichment and identification of >100 endogenous proteins, including many not previously linked to endoplasmic reticulum-mitochondria contacts. We validated eight candidates by biochemical fractionation and overexpression imaging. Overall, split-TurboID is a versatile tool for conditional and spatially specific proximity labeling in cells.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteoma/análisis , Biotinilación , Células HEK293 , Humanos , Proteoma/metabolismo , Coloración y Etiquetado
7.
Mol Cell ; 55(2): 332-41, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25002142

RESUMEN

Obtaining complete protein inventories for subcellular regions is a challenge that often limits our understanding of cellular function, especially for regions that are impossible to purify and are therefore inaccessible to traditional proteomic analysis. We recently developed a method to map proteomes in living cells with an engineered peroxidase (APEX) that bypasses the need for organellar purification when applied to membrane-bound compartments; however, it was insufficiently specific when applied to unbounded regions that allow APEX-generated radicals to escape. Here, we combine APEX technology with a SILAC-based ratiometric tagging strategy to substantially reduce unwanted background and achieve nanometer spatial resolution. This is applied to map the proteome of the mitochondrial intermembrane space (IMS), which can freely exchange small molecules with the cytosol. Our IMS proteome of 127 proteins has >94% specificity and includes nine newly discovered mitochondrial proteins. This approach will enable scientists to map proteomes of cellular regions that were previously inaccessible.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Proteoma/metabolismo , Western Blotting , Fraccionamiento Celular , Células HEK293 , Humanos , Marcaje Isotópico , Membranas Mitocondriales/metabolismo
8.
Nature ; 523(7559): 183-188, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26131937

RESUMEN

Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). Here, we demonstrate that lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1α) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)), resulting in CK1α degradation. CK1α is encoded by a gene within the common deleted region for del(5q) MDS and haploinsufficient expression sensitizes cells to lenalidomide therapy, providing a mechanistic basis for the therapeutic window of lenalidomide in del(5q) MDS. We found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degradation of CK1α. We further demonstrate that minor side chain modifications in thalidomide and a novel analogue, CC-122, can modulate the spectrum of substrates targeted by CRL4(CRBN). These findings have implications for the clinical activity of lenalidomide and related compounds, and demonstrate the therapeutic potential of novel modulators of E3 ubiquitin ligases.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/fisiopatología , Talidomida/análogos & derivados , Ubiquitinación/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Quinasa de la Caseína I/genética , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Factores Inmunológicos/farmacología , Células Jurkat , Células K562 , Lenalidomida , Ratones , Datos de Secuencia Molecular , Péptido Hidrolasas/química , Proteolisis/efectos de los fármacos , Alineación de Secuencia , Eliminación de Secuencia , Especificidad de la Especie , Talidomida/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
9.
Nat Methods ; 14(12): 1167-1170, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29039416

RESUMEN

Although purification of biotinylated molecules is highly efficient, identifying specific sites of biotinylation remains challenging. We show that anti-biotin antibodies enable unprecedented enrichment of biotinylated peptides from complex peptide mixtures. Live-cell proximity labeling using APEX peroxidase followed by anti-biotin enrichment and mass spectrometry yielded over 1,600 biotinylation sites on hundreds of proteins, an increase of more than 30-fold in the number of biotinylation sites identified compared to streptavidin-based enrichment of proteins.


Asunto(s)
Anticuerpos/metabolismo , Biotina/metabolismo , Péptidos/química , Proteínas/química , Biotecnología/métodos , Biotinilación , Cromatografía Liquida , Células HEK293 , Humanos , Células Jurkat , Proteínas/aislamiento & purificación , Coloración y Etiquetado , Estreptavidina/metabolismo , Espectrometría de Masas en Tándem
10.
Blood ; 132(14): 1535-1544, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30064974

RESUMEN

Thalidomide and its derivatives, lenalidomide and pomalidomide, are clinically effective treatments for multiple myeloma and myelodysplastic syndrome with del(5q). These molecules lack activity in murine models, limiting investigation of their therapeutic activity or toxicity in vivo. Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). In human cells, thalidomide and its analogs bind CRBN and recruit protein targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and subsequent degradation by the proteasome. We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1α), both in vitro and in vivo. We use the Crbn I391V model to demonstrate that the in vivo therapeutic activity of lenalidomide in del(5q) myelodysplastic syndrome can be explained by heterozygous expression of Ck1α in del(5q) cells. We found that lenalidomide acts on hematopoietic stem cells with heterozygous expression of Ck1α and inactivation of Trp53 causes lenalidomide resistance. We further demonstrate that Crbn I391V is sufficient to confer thalidomide-induced fetal loss in mice, capturing a major toxicity of this class of drugs. Further study of the Crbn I391V model will provide valuable insights into the in vivo efficacy and toxicity of this class of drugs.


Asunto(s)
Antineoplásicos/farmacología , Lenalidomida/farmacología , Síndromes Mielodisplásicos/tratamiento farmacológico , Proteínas del Tejido Nervioso/genética , Mutación Puntual , Talidomida/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antineoplásicos/química , Quinasa de la Caseína I/metabolismo , Modelos Animales de Enfermedad , Femenino , Hematopoyesis/efectos de los fármacos , Lenalidomida/química , Masculino , Ratones , Ratones Endogámicos C57BL , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Talidomida/análogos & derivados
11.
Mol Cell Proteomics ; 14(9): 2429-40, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25953088

RESUMEN

Introduction of antibodies specific for acetylated lysine has significantly improved the detection of endogenous acetylation sites by mass spectrometry. Here, we describe a new, commercially available mixture of anti-lysine acetylation (Kac) antibodies and show its utility for in-depth profiling of the acetylome. Specifically, seven complementary monoclones with high specificity for Kac were combined into a final anti-Kac reagent which results in at least a twofold increase in identification of Kac peptides over a commonly used Kac antibody. We outline optimal antibody usage conditions, effective offline basic reversed phase separation, and use of state-of-the-art LC-MS technology for achieving unprecedented coverage of the acetylome. The methods were applied to quantify acetylation sites in suberoylanilide hydroxamic acid-treated Jurkat cells. Over 10,000 Kac peptides from over 3000 Kac proteins were quantified from a single stable isotope labeling by amino acids in cell culture labeled sample using 7.5 mg of peptide input per state. This constitutes the deepest coverage of acetylation sites in quantitative experiments obtained to-date. The approach was also applied to breast tumor xenograft samples using isobaric mass tag labeling of peptides (iTRAQ4, TMT6 and TMT10-plex reagents) for quantification. Greater than 6700 Kac peptides from over 2300 Kac proteins were quantified using 1 mg of tumor protein per iTRAQ 4-plex channel. The novel reagents and methods we describe here enable quantitative, global acetylome analyses with depth and sensitivity approaching that obtained for other well-studied post-translational modifications such as phosphorylation and ubiquitylation, and should have widespread application in biological and clinical studies employing mass spectrometry-based proteomics.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Hígado/metabolismo , Lisina/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Proteómica/métodos , Acetilación , Animales , Femenino , Humanos , Células Jurkat , Lisina/inmunología , Espectrometría de Masas/métodos , Ratones , Procesamiento Proteico-Postraduccional , Flujo de Trabajo
12.
Mol Cell Proteomics ; 12(3): 825-31, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23266961

RESUMEN

Detection of endogenous ubiquitination sites by mass spectrometry has dramatically improved with the commercialization of anti-di-glycine remnant (K-ε-GG) antibodies. Here, we describe a number of improvements to the K-ε-GG enrichment workflow, including optimized antibody and peptide input requirements, antibody cross-linking, and improved off-line fractionation prior to enrichment. This refined and practical workflow enables routine identification and quantification of ∼20,000 distinct endogenous ubiquitination sites in a single SILAC experiment using moderate amounts of protein input.


Asunto(s)
Proteoma/análisis , Proteómica/métodos , Ubiquitinación , Aminoácidos/metabolismo , Anticuerpos/química , Anticuerpos/inmunología , Sitios de Unión , Cromatografía Liquida/métodos , Reactivos de Enlaces Cruzados/química , Inhibidores de Cisteína Proteinasa/farmacología , Glicilglicina/inmunología , Humanos , Marcaje Isotópico/métodos , Células Jurkat , Leupeptinas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteoma/química , Proteoma/metabolismo , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem/métodos , Proteínas Ubiquitinadas/análisis , Proteínas Ubiquitinadas/química , Proteínas Ubiquitinadas/metabolismo
13.
J Cell Biol ; 223(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38578285

RESUMEN

IRE1α is an endoplasmic reticulum (ER) sensor that recognizes misfolded proteins to induce the unfolded protein response (UPR). We studied cholera toxin (CTx), which invades the ER and activates IRE1α in host cells, to understand how unfolded proteins are recognized. Proximity labeling colocalized the enzymatic and metastable A1 segment of CTx (CTxA1) with IRE1α in live cells, where we also found that CTx-induced IRE1α activation enhanced toxicity. In vitro, CTxA1 bound the IRE1α lumenal domain (IRE1αLD), but global unfolding was not required. Rather, the IRE1αLD recognized a seven-residue motif within an edge ß-strand of CTxA1 that must locally unfold for binding. Binding mapped to a pocket on IRE1αLD normally occupied by a segment of the IRE1α C-terminal flexible loop implicated in IRE1α oligomerization. Mutation of the CTxA1 recognition motif blocked CTx-induced IRE1α activation in live cells, thus linking the binding event with IRE1α signal transduction and induction of the UPR.


Asunto(s)
Toxina del Cólera , Endorribonucleasas , Proteínas Serina-Treonina Quinasas , Respuesta de Proteína Desplegada , Toxina del Cólera/genética , Toxina del Cólera/metabolismo , Estrés del Retículo Endoplásmico , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Humanos , Animales , Ratones , Línea Celular
14.
bioRxiv ; 2023 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-37745534

RESUMEN

PCP signaling polarizes epithelial cells within the plane of an epithelium. Core PCP signaling components adopt asymmetric subcellular localizations within cells to both polarize and coordinate polarity between cells. Achieving subcellular asymmetry requires additional effectors, including some mediating post-translational modifications of core components. Identification of such proteins is challenging due to pleiotropy. We used mass spectrometry-based proximity labeling proteomics to identify such regulators in the Drosophila wing. We identified the catalytic subunit of Protein Phosphatase1, Pp1-87B, and show that it regulates core protein polarization. Pp1-87B interacts with the core protein Van Gogh and at least one Serine/Threonine kinase, Dco/CKIε, that is known to regulate PCP. Pp1-87B modulates Van Gogh subcellular localization and directs its dephosphorylation in vivo. PNUTS, a Pp1 regulatory subunit, also modulates PCP. While the direct substrate(s) of Pp1-87B in control of PCP is not known, our data support the model that cycling between phosphorylated and unphosphorylated forms of one or more core PCP components may regulate acquisition of asymmetry. Finally, our screen serves as a resource for identifying additional regulators of PCP signaling.

15.
Neuron ; 110(14): 2299-2314.e8, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35613619

RESUMEN

Transcription factors specify the fate and connectivity of developing neurons. We investigate how a lineage-specific transcription factor, Acj6, controls the precise dendrite targeting of Drosophila olfactory projection neurons (PNs) by regulating the expression of cell-surface proteins. Quantitative cell-surface proteomic profiling of wild-type and acj6 mutant PNs in intact developing brains, and a proteome-informed genetic screen identified PN surface proteins that execute Acj6-regulated wiring decisions. These include canonical cell adhesion molecules and proteins previously not associated with wiring, such as Piezo, whose mechanosensitive ion channel activity is dispensable for its function in PN dendrite targeting. Comprehensive genetic analyses revealed that Acj6 employs unique sets of cell-surface proteins in different PN types for dendrite targeting. Combined expression of Acj6 wiring executors rescued acj6 mutant phenotypes with higher efficacy and breadth than expression of individual executors. Thus, Acj6 controls wiring specificity of different neuron types by specifying distinct combinatorial expression of cell-surface executors.


Asunto(s)
Proteínas de Drosophila , Neuronas Receptoras Olfatorias , Animales , Dendritas/fisiología , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Canales Iónicos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vías Olfatorias/fisiología , Neuronas Receptoras Olfatorias/metabolismo , Factores del Dominio POU/metabolismo , Proteómica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Nat Commun ; 12(1): 2382, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888706

RESUMEN

Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteómica/métodos , Proteínas Represoras/metabolismo , Coloración y Etiquetado/métodos , Animales , Animales Modificados Genéticamente , Biotina/metabolismo , Biotinilación , Ligasas de Carbono-Nitrógeno/genética , Línea Celular , Modelos Animales de Enfermedad , Drosophila , Células Madre Embrionarias , Proteínas de Escherichia coli/genética , Femenino , Humanos , Masculino , Ratones , Ingeniería de Proteínas , Transporte de Proteínas , Proteínas Represoras/genética , Espectrometría de Masas en Tándem/métodos , Teratoma/diagnóstico , Teratoma/patología
17.
Nat Commun ; 12(1): 734, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531470

RESUMEN

Driver genes with a mutually exclusive mutation pattern across tumor genomes are thought to have overlapping roles in tumorigenesis. In contrast, we show here that mutually exclusive prostate cancer driver alterations involving the ERG transcription factor and the ubiquitin ligase adaptor SPOP are synthetic sick. At the molecular level, the incompatible cancer pathways are driven by opposing functions in SPOP. ERG upregulates wild type SPOP to dampen androgen receptor (AR) signaling and sustain ERG activity through degradation of the bromodomain histone reader ZMYND11. Conversely, SPOP-mutant tumors stabilize ZMYND11 to repress ERG-function and enable oncogenic androgen receptor signaling. This dichotomy regulates the response to therapeutic interventions in the AR pathway. While mutant SPOP renders tumor cells susceptible to androgen deprivation therapies, ERG promotes sensitivity to high-dose androgen therapy and pharmacological inhibition of wild type SPOP. More generally, these results define a distinct class of antagonistic cancer drivers and a blueprint toward their therapeutic exploitation.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Represoras/metabolismo , Regulador Transcripcional ERG/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Desnudos , Mutación/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Neoplasias de la Próstata/genética , Unión Proteica , Proteómica , Receptores Androgénicos/metabolismo , Proteínas Represoras/genética , Transducción de Señal/fisiología , Regulador Transcripcional ERG/genética , Complejos de Ubiquitina-Proteína Ligasa/genética
18.
Nat Commun ; 11(1): 359, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31953384

RESUMEN

Protein ubiquitylation is involved in a plethora of cellular processes. While antibodies directed at ubiquitin remnants (K-ɛ-GG) have improved the ability to monitor ubiquitylation using mass spectrometry, methods for highly multiplexed measurement of ubiquitylation in tissues and primary cells using sub-milligram amounts of sample remains a challenge. Here, we present a highly sensitive, rapid and multiplexed protocol termed UbiFast for quantifying ~10,000 ubiquitylation sites from as little as 500 µg peptide per sample from cells or tissue in a TMT10plex in ca. 5 h. High-field Asymmetric Waveform Ion Mobility Spectrometry (FAIMS) is used to improve quantitative accuracy for posttranslational modification analysis. We use the approach to rediscover substrates of the E3 ligase targeting drug lenalidomide and to identify proteins modulated by ubiquitylation in models of basal and luminal human breast cancer. The sensitivity and speed of the UbiFast method makes it suitable for large-scale studies in primary tissue samples.


Asunto(s)
Proteínas/metabolismo , Proteoma/análisis , Investigación Biomédica Traslacional/métodos , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Animales , Neoplasias de la Mama , Caseína Quinasa Ialfa , Femenino , Células HeLa , Humanos , Factor de Transcripción Ikaros , Espectrometría de Masas/métodos , Ratones , Mieloma Múltiple , Procesamiento Proteico-Postraduccional , Proteómica/métodos , Sensibilidad y Especificidad , Coloración y Etiquetado , Ubiquitina-Proteína Ligasas/metabolismo
19.
Nat Biotechnol ; 38(1): 108, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31748691

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

20.
Mol Cancer Res ; 18(4): 574-584, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31862696

RESUMEN

Genomic analysis of lung adenocarcinomas has revealed that the MGA gene, which encodes a heterodimeric partner of the MYC-interacting protein MAX, is significantly mutated or deleted in lung adenocarcinomas. Most of the mutations are loss of function for MGA, suggesting that MGA may act as a tumor suppressor. Here, we characterize both the molecular and cellular role of MGA in lung adenocarcinomas and illustrate its functional relevance in the MYC pathway. Although MGA and MYC interact with the same binding partner, MAX, and recognize the same E-box DNA motif, we show that the molecular function of MGA appears to be antagonistic to that of MYC. Using mass spectrometry-based affinity proteomics, we demonstrate that MGA interacts with a noncanonical PCGF6-PRC1 complex containing MAX and E2F6 that is involved in gene repression, while MYC is not part of this MGA complex, in agreement with previous studies describing the interactomes of E2F6 and PCGF6. Chromatin immunoprecipitation-sequencing and RNA sequencing assays show that MGA binds to and represses genes that are bound and activated by MYC. In addition, we show that, as opposed to the MYC oncoprotein, MGA acts as a negative regulator for cancer cell proliferation. Our study defines a novel MYC/MAX/MGA pathway, in which MYC and MGA play opposite roles in protein interaction, transcriptional regulation, and cellular proliferation. IMPLICATIONS: This study expands the range of key cancer-associated genes whose dysregulation is functionally equivalent to MYC activation and places MYC within a linear pathway analogous to cell-cycle or receptor tyrosine kinase/RAS/RAF pathways in lung adenocarcinomas.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células A549 , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Proteínas Proto-Oncogénicas c-myc/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA