Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Bioconjug Chem ; 28(7): 1906-1915, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28590752

RESUMEN

Phosphopantetheine transferases (PPTases) can be used to efficiently prepare site-specific antibody-drug conjugates (ADCs) by enzymatically coupling coenzyme A (CoA)-linker payloads to 11-12 amino acid peptide substrates inserted into antibodies. Here, a two-step strategy is established wherein in a first step, CoA analogs with various bioorthogonal reactivities are enzymatically installed on the antibody for chemical conjugation with a cytotoxic payload in a second step. Because of the high structural similarity of these CoA analogs to the natural PPTase substrate CoA-SH, the first step proceeds very efficiently and enables the use of peptide tags as short as 6 amino acids compared to the 11-12 amino acids required for efficient one-step coupling of the payload molecule. Furthermore, two-step conjugation provides access to diverse linker chemistries and spacers of varying lengths. The potency of the ADCs was largely independent of linker architecture. In mice, proteolytic cleavage was observed for some C-terminally linked auristatin payloads. The in vivo stability of these ADCs was significantly improved by reduction of the linker length. In addition, linker stability was found to be modulated by attachment site, and this, together with linker length, provides an opportunity for maximizing ADC stability without sacrificing potency.


Asunto(s)
Anticuerpos Monoclonales/química , Coenzima A/química , Citotoxinas/química , Inmunoconjugados/química , Aminobenzoatos/administración & dosificación , Aminobenzoatos/química , Animales , Citotoxinas/administración & dosificación , Estabilidad de Medicamentos , Ratones , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Relación Estructura-Actividad
2.
Mol Biosyst ; 12(6): 1756-9, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27063346

RESUMEN

The post-translational modification of serine or threonine residues of proteins with a single N-acetylglucosamine monosaccharide (O-GlcNAcylation) is essential for cell survival and function. However, relatively few O-GlcNAc modification sites have been mapped due to the difficulty of enriching and detecting O-GlcNAcylated peptides from complex samples. Here we describe an improved approach to quantitatively label and enrich O-GlcNAcylated proteins for site identification. Chemoenzymatic labelling followed by copper(i)-catalysed azide-alkyne cycloaddition (CuAAC) installs a new mass spectrometry (MS)-compatible linker designed for facile purification of O-GlcNAcylated proteins from cell lysates. The linker also allows subsequent quantitative release of O-GlcNAcylated proteins for downstream MS analysis. We validate the approach by unambiguously identifying several established O-GlcNAc sites on the proteins α-crystallin and O-GlcNAc transferase (OGT), as well as discovering new, previously unreported sites on OGT. Notably, these novel sites on OGT lie in key functional domains of the protein, underscoring how this site identification method may reveal important biological insights into protein activity and regulation.


Asunto(s)
Acetilglucosamina/química , Acetilglucosamina/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional , Cromatografía Liquida , Glicosilación , Humanos , Espectrometría de Masas , Coloración y Etiquetado
3.
Cell ; 164(3): 499-511, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824658

RESUMEN

The volume-regulated anion channel (VRAC) is activated when a cell swells, and it plays a central role in maintaining cell volume in response to osmotic challenges. SWELL1 (LRRC8A) was recently identified as an essential component of VRAC. However, the identity of the pore-forming subunits of VRAC and how the channel is gated by cell swelling are unknown. Here, we show that SWELL1 and up to four other LRRC8 subunits assemble into heterogeneous complexes of ∼800 kDa. When reconstituted into bilayers, LRRC8 complexes are sufficient to form anion channels activated by osmolality gradients. In bilayers, as well as in cells, the single-channel conductance of the complexes depends on the LRRC8 composition. Finally, low ionic strength (Γ) in the absence of an osmotic gradient activates the complexes in bilayers. These data demonstrate that LRRC8 proteins together constitute the VRAC pore and that hypotonic stress can activate VRAC through a decrease in cytoplasmic Γ.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas de la Membrana/metabolismo , Células HeLa , Humanos , Canales Iónicos/química , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Ósmosis
4.
J Immunol ; 194(6): 2539-50, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25672758

RESUMEN

Rheumatoid arthritis (RA) is associated with amino acid variants in multiple MHC molecules. The association to MHC class II (MHC-II) has been studied in several animal models of RA. In most cases these models depend on T cells restricted to a single immunodominant peptide of the immunizing Ag, which does not resemble the autoreactive T cells in RA. An exception is pristane-induced arthritis (PIA) in the rat where polyclonal T cells induce chronic arthritis after being primed against endogenous Ags. In this study, we used a mixed genetic and functional approach to show that RT1-Ba and RT1-Bb (RT1-B locus), the rat orthologs of HLA-DQA and HLA-DQB, determine the onset and severity of PIA. We isolated a 0.2-Mb interval within the MHC-II locus of three MHC-congenic strains, of which two were protected from severe PIA. Comparison of sequence and expression variation, as well as in vivo blocking of RT1-B and RT1-D (HLA-DR), showed that arthritis in these strains is regulated by coding polymorphisms in the RT1-B genes. Motif prediction based on MHC-II eluted peptides and structural homology modeling suggested that variants in the RT1-B P1 pocket, which likely affect the editing capacity by RT1-DM, are important for the development of PIA.


Asunto(s)
Artritis Experimental/genética , Artritis Reumatoide/genética , Predisposición Genética a la Enfermedad , Antígenos de Histocompatibilidad/genética , Secuencia de Aminoácidos , Aminoácidos/genética , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Bloqueadores/farmacología , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Sitios de Unión/genética , Peso Corporal/efectos de los fármacos , Peso Corporal/inmunología , Modelos Animales de Enfermedad , Genotipo , Haplotipos/inmunología , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/inmunología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Polimorfismo Genético/inmunología , Estructura Terciaria de Proteína , Ratas , Índice de Severidad de la Enfermedad , Terpenos/inmunología
5.
Cell Rep ; 9(2): 495-503, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25373897

RESUMEN

Hippo signaling is a tumor-suppressor pathway involved in organ size control and tumorigenesis through the inhibition of YAP and TAZ. Here, we show that energy stress induces YAP cytoplasmic retention and S127 phosphorylation and inhibits YAP transcriptional activity and YAP-dependent transformation. These effects require the central metabolic sensor AMP-activated protein kinase (AMPK) and the upstream Hippo pathway components Lats1/Lats2 and angiomotin-like 1 (AMOTL1). Furthermore, we show that AMPK directly phosphorylates S793 of AMOTL1. AMPK activation stabilizes and increases AMOTL1 steady-state protein levels, contributing to YAP inhibition. The phosphorylation-deficient S793Ala mutant of AMOTL1 showed a shorter half-life and conferred resistance to energy-stress-induced YAP inhibition. Our findings link energy sensing to the Hippo-YAP pathway and suggest that YAP may integrate spatial (contact inhibition), mechanical, and metabolic signals to control cellular proliferation and survival.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenilato Quinasa/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Estrés Fisiológico , Secuencia de Aminoácidos , Angiomotinas , Metabolismo Energético , Células HEK293 , Vía de Señalización Hippo , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Mutación Missense , Fosforilación , Estabilidad Proteica , Factores de Transcripción , Proteínas Supresoras de Tumor/metabolismo , Proteínas Señalizadoras YAP
6.
J Am Chem Soc ; 136(27): 9528-31, 2014 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-24937314

RESUMEN

Although fucose-α(1-2)-galactose (Fucα(1-2)Gal)-containing glycans have been implicated in cognitive processes such as learning and memory, their precise molecular mechanisms are poorly understood. Here we employed the use of multivalent glycopolymers to enable the first proteome-wide identification of weak affinity, low abundance Fucα(1-2)Gal glycan-binding proteins (GBPs). Biotin-terminated glycopolymers containing photoactivatable cross-linking groups were designed to capture and enrich GBPs from rat brain lysates. Candidate proteins were tested for their ability to bind Fucα(1-2)Gal, and the functional significance of the interaction was investigated for the synaptic vesicle protein SV2a using a knockout mouse system. The results suggest a role for SV2a-Fucα(1-2)Gal interactions in SV2a trafficking and synaptic vesicle recycling. More broadly, our studies outline a general chemical approach for the systems-level discovery of novel GBPs.


Asunto(s)
Galactosa/química , Polisacáridos/metabolismo , Polisacáridos/efectos de la radiación , Proteoma/análisis , Proteoma/metabolismo , Animales , Encéfalo/metabolismo , Galactosa/análogos & derivados , Ratones , Ratones Noqueados , Estructura Molecular , Procesos Fotoquímicos , Proteoma/química , Ratas
7.
FASEB J ; 28(8): 3758-68, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24823363

RESUMEN

Citrullinated collagen II (CII) is a well-known autoantigen in rheumatoid arthritis (RA). However, the direct effects of CII citrullination on cell behavior have not been described. To study whether citrullination of CII could affect cellular functions, we measured the adhesion of 3 different cell types (human Saos2 osteosarcoma cells, human synovial fibroblasts, and rat mesenchymal stem cells) with impedance-based technology. The binding of different collagen receptor integrins to citrullinated collagen was studied by CHO cell lines, each overexpressing 1 of the 4 human collagen receptors on the cell surface, and with solid-phase binding assays, using the recombinant human integrin α1I, α2I, α10I, and α11I domains. Collagen citrullination decreased the adhesion of synovial fibroblasts ∼50% (P<0.05) and mesenchymal stem cells ∼40% (P<0.05) by specifically decreasing the binding of integrins α10ß1 and α11ß1 to arginine-containing motifs, such as GFOGER. In contrast, citrullination had only a minor effect on the function of α1ß1 and α2ß1 integrins, which have been reported to play a critical role in regulating leukocyte function. Molecular modeling was used to explain the detected functional differences at the structural level. Given that the integrins regulate cell metabolism, proliferation, and migration, we suggest that collagen citrullination modifies the pathogenesis of RA. Here, CII citrullination was shown to decrease the survival of mesenchymal stem cells.


Asunto(s)
Adhesión Celular/fisiología , Citrulina/química , Colágeno Tipo II/química , Integrinas/fisiología , Secuencias de Aminoácidos , Aminoacilación , Animales , Arginina/química , Neoplasias Óseas/patología , Células CHO , Línea Celular Tumoral , Células Cultivadas , Pollos , Colágeno Tipo II/farmacología , Cricetinae , Cricetulus , Fibroblastos/patología , Humanos , Integrinas/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoartritis/patología , Osteosarcoma/patología , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Membrana Sinovial/patología , Transfección
8.
Arthritis Rheumatol ; 66(6): 1440-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24470447

RESUMEN

OBJECTIVE: To investigate type II collagen (CII) as a joint-specific target of the anti-citrullinated protein antibody (ACPA) response in rheumatoid arthritis (RA). METHODS: Potential citrullinated neoepitopes were identified by high-resolution tandem mass spectrometry (MS/MS) of in vitro peptidylarginine deiminase 2 (PAD-2)-treated CII, and the relationship between citrullination and CII conformation was investigated by circular dichroism and conformation-dependent antibodies. Based on the MS analyses, synthetic peptides were designed and analyzed for serum IgG reactivity in the Epidemiological Investigation of RA (EIRA) case-control cohort of 1,949 RA patients and 278 healthy controls. Peptide-specific antibodies were purified from RA patient serum and used to stain RA cartilage specimens. RESULTS: We described the conformation-dependent citrullination pattern of CII after PAD-2 treatment at room temperature and 37°C and showed that CII could be citrullinated in its native triple-helical conformation. Screening of Arg and Cit pairs of synthetic peptides revealed new citrullinated B cell epitopes on CII. Antibodies directed to 2 proximal epitopes close to the C-terminus of the CII triple helix were recognized by autoantibodies in 21% and 17% of RA patients, respectively. Affinity-purified antibodies from RA sera directed to these 2 epitopes, but not antibodies directed to citrullinated α-enolase peptide 1, bound to RA cartilage. CONCLUSION: These findings suggest that cartilage-directed anticitrulline immunity contributes to the induction of joint inflammation in RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Autoanticuerpos/metabolismo , Cartílago Articular/metabolismo , Citrulina/inmunología , Colágeno Tipo II/metabolismo , Animales , Estudios de Casos y Controles , Colágeno Tipo II/efectos de los fármacos , Epítopos/inmunología , Humanos , Hidrolasas/farmacología , Inmunoglobulina G/metabolismo , Técnicas In Vitro , Desiminasas de la Arginina Proteica , Ratas , Espectrometría de Masas en Tándem
9.
Sci Signal ; 6(290): ra75, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23982206

RESUMEN

The transcription factor nuclear factor κB (NF-κB) rapidly reprograms gene expression in response to various stimuli, and its activity is regulated by several posttranslational modifications, including phosphorylation, methylation, and acetylation. The addition of O-linked ß-N-acetylglucosamine (a process known as O-GlcNAcylation) is an abundant posttranslational modification that is enhanced in conditions such as hyperglycemia and cellular stress. We report that the NF-κB subunit c-Rel is modified and activated by O-GlcNAcylation. We identified serine 350 as the site of O-GlcNAcylation, which was required for the DNA binding and transactivation functions of c-Rel. Blocking the O-GlcNAcylation of this residue abrogated c-Rel-mediated expression of the cytokine-encoding genes IL2, IFNG, and CSF2 in response to T cell receptor (TCR) activation, whereas increasing the extent of O-GlcNAcylation of cellular proteins enhanced the expression of these genes. TCR- or tumor necrosis factor (TNF)-induced expression of other NF-κB target genes, such as NFKBIA (which encodes IκBα) and TNFAIP3 (which encodes A20), occurred independently of the O-GlcNAcylation of c-Rel. Our findings suggest a stimulus-specific role for hyperglycemia-induced O-GlcNAcylation of c-Rel in promoting T cell-mediated autoimmunity in conditions such as type 1 diabetes by enhancing the production of T helper cell cytokines.


Asunto(s)
Acetilglucosamina/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Acilación , Animales , Sitios de Unión/genética , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Glicosilación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células HEK293 , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , FN-kappa B/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-rel/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina/genética , Serina/metabolismo , Transducción de Señal
10.
ACS Chem Biol ; 8(8): 1764-74, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23773140

RESUMEN

The cell utilizes the Keap1/Nrf2-ARE signaling pathway to detoxify harmful chemicals in order to protect itself from oxidative stress and to maintain its reducing environment. When exposed to oxidative stress and xenobiotic inducers, the redox sensitive Keap1 is covalently modified at specific cysteine residues. Consequently, the latent transcription factor Nrf2 is stabilized and translocates into the nucleus, where it transactivates the expression of detoxification genes through binding to the antioxidant response element (ARE). In the pursuit of potent and bioavailable activators of the ARE, we validated hits from a pathway-directed high-throughput screening campaign by testing them in cell culture and a reporter strain of a whole animal model, Caenorhabditis elegans. These studies allowed us to identify AI-3 as an ARE activator that induces cytoprotective genes in human cells and in worms, which also translated into in vivo activity in mice. AI-3 is an electrophilic ARE activator with two thiol sensitive sites toward a nucleophilic aromatic substitution, and SAR studies indicated the tunability of the system. Tandem LC-MS analysis revealed that AI-3 alkylates Keap1 primarily at Cys151, while AI-3 is reactive toward additional cysteine residues at higher doses in vitro and in vivo. The immediate effects of such alkylation included the disruption of Keap1-Cul3 (low [AI-3]) and/or Keap1-Nrf2 (high [AI-3]) interactions that both led to the stabilization of Nrf2. This further translated into the downstream Nrf2-ARE regulated cytoprotective gene activation. Collectively, AI-3 may become a valuable biological tool and may even provide therapeutic benefits in oxidative stress related diseases.


Asunto(s)
Hidrolasas de Éster Carboxílico/química , Sondas Moleculares/química , Factor 2 Relacionado con NF-E2/química , Fosfatidilinositol 3-Quinasas/química , Sulfonas/química , Tiofenos/química , Animales , Caenorhabditis elegans/química , Línea Celular , Células Cultivadas , Cromatografía Liquida , Humanos , Ratones , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Sulfonas/farmacología , Tiofenos/farmacología
11.
ACS Chem Biol ; 8(7): 1407-11, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23621521

RESUMEN

Retinal pigment epithelial (RPE) cells form a monolayer adjacent to the retina and play a critical role in the visual light cycle. Degeneration of RPE cells results in retinal disorders such as age-related macular degeneration. Cell transplant strategies have potential therapeutic value for such disorders; however, risks associated with an inadequate supply of donor cells limit their therapeutic success. The identification of factors that proliferate RPE cells ex vivo could provide a renewable source of cells for transplantation. Here, we report that a small molecule (WS3) can reversibly proliferate primary RPE cells isolated from fetal and adult human donors. Following withdrawal of WS3, RPE cells differentiate into a functional monolayer, as exhibited by their expression of mature RPE genes and phagocytosis of photoreceptor outer segments. Furthermore, chemically expanded RPE cells preserve vision when transplanted into dystrophic Royal College of Surgeons (RCS) rats, a well-established model of retinal degeneration.


Asunto(s)
Biotina/análogos & derivados , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Pirimidinas/farmacología , Retina/citología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Biotina/química , Biotina/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Madre Fetales , Técnica del Anticuerpo Fluorescente , Humanos , Estructura Molecular , Compuestos de Fenilurea/química , Pirimidinas/química , Ratas , Retina/efectos de los fármacos , Degeneración Retiniana/tratamiento farmacológico
12.
J Neurosci ; 33(6): 2457-64, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23392674

RESUMEN

Accumulation of ß-amyloid (Aß) in the brain is believed to contribute to the pathology of Alzheimer's Disease (AD). Aß levels are controlled by the production of Aß from amyloid precursor protein, degradation by proteases, and peripheral clearance. In this study we sought to determine whether enhancing clearance of plasma Aß with a peripherally administered Aß-degrading protease would reduce brain Aß levels through a peripheral sink. Neprilysin (NEP) is a zinc-dependent metalloprotease that is one of the key Aß-degrading enzymes in the brain. We developed a NEP fusion protein with in vitro degradation of Aß and a 10 day plasma half-life in mouse. Intravenous administration of NEP to wild-type and APP23 transgenic mice resulted in dose-dependent clearance of plasma Aß. However, this did not correspond to reduced levels of soluble brain Aß with treatment up to 5 weeks in WT mice or formic acid-extractable brain Aß with 3 month treatment in aged APP23. In contrast, intracranial injection of NEP resulted in an acute decrease in soluble brain Aß. We found no change in amyloid precursor protein gene expression in mice treated with intravenous NEP, suggesting that the lack of effects in the brain following this route of administration was not caused by compensatory upregulation of Aß production. Taken together, these results suggest a lack of a robust peripheral Aß efflux sink through which brain amyloid burdens can be therapeutically reduced.


Asunto(s)
Péptidos beta-Amiloides/sangre , Precursor de Proteína beta-Amiloide/sangre , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Neprilisina/farmacología , Proteolisis/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular
13.
Science ; 337(6097): 975-80, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22923583

RESUMEN

Cancer cells must satisfy the metabolic demands of rapid cell growth within a continually changing microenvironment. We demonstrated that the dynamic posttranslational modification of proteins by O-linked ß-N-acetylglucosamine (O-GlcNAcylation) is a key metabolic regulator of glucose metabolism. O-GlcNAcylation was induced at serine 529 of phosphofructokinase 1 (PFK1) in response to hypoxia. Glycosylation inhibited PFK1 activity and redirected glucose flux through the pentose phosphate pathway, thereby conferring a selective growth advantage on cancer cells. Blocking glycosylation of PFK1 at serine 529 reduced cancer cell proliferation in vitro and impaired tumor formation in vivo. These studies reveal a previously uncharacterized mechanism for the regulation of metabolic pathways in cancer and a possible target for therapeutic intervention.


Asunto(s)
Proliferación Celular , Glucosa/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fosfofructoquinasa-1 Tipo Hepático/metabolismo , Acetilglucosamina/metabolismo , Acilación , Adenosina Trifosfato/metabolismo , Animales , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Glucólisis , Glicosilación , Humanos , Ácido Láctico/metabolismo , Ratones , Ratones Desnudos , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , NADP/metabolismo , Vía de Pentosa Fosfato , Fosfofructoquinasa-1 Tipo Hepático/antagonistas & inhibidores , Fosfofructoquinasa-1 Tipo Hepático/química
14.
Chem Biol ; 19(8): 955-62, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22884261

RESUMEN

Hippo signaling represents a tumor suppressor pathway that regulates organ size and tumorigenesis through phosphorylation and inhibition of the transcription coactivator YAP. Here, we show that serum deprivation dramatically induces YAP Ser127 phosphorylation and cytoplasmic retention, independent of cell-cell contact. Through chemical isolation and activity profiling, we identified serum-derived sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) as small molecule activators of YAP. S1P induces YAP nuclear localization through S1P(2) receptor, Rho GTPase activation, and F-actin polymerization, independent of the core Hippo pathway kinases. Bioinformatics studies also showed that S1P stimulation induces YAP target gene expression in mouse liver and human embryonic stem cells. These results revealed potent small molecule regulators of YAP and suggest that S1P and LPA might modulate cell proliferation and tumorigenesis through YAP activation.


Asunto(s)
Lisofosfolípidos/farmacología , Proteínas Nucleares/metabolismo , Esfingosina/análogos & derivados , Factores de Transcripción/metabolismo , Actinas/metabolismo , Animales , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Lisofosfolípidos/sangre , Lisofosfolípidos/química , Lisofosfolípidos/aislamiento & purificación , Ratones , Proteínas Nucleares/química , Fosforilación/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/sangre , Esfingosina/aislamiento & purificación , Esfingosina/farmacología , Factores de Transcripción/química , Proteínas de Unión al GTP rho/metabolismo
15.
J Neurosci ; 32(20): 6894-905, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22593058

RESUMEN

Local control of calcium concentration within neurons is critical for signaling and regulation of synaptic communication in neural circuits. How local control can be achieved in the absence of physical compartmentalization is poorly understood. Challenging examples are provided by nicotinic acetylcholine receptors that contain α7 nicotinic receptor subunits (α7-nAChRs). These receptors are highly permeable to calcium and are concentrated on aspiny dendrites of interneurons, which lack obvious physical compartments for constraining calcium diffusion. Using functional proteomics on rat brain, we show that α7-nAChRs are associated with plasma membrane calcium-ATPase pump isoform 2 (PMCA2). Analysis of α7-nAChR function in hippocampal interneurons in culture shows that PMCA2 activity limits the duration of calcium elevations produced by the receptors. Unexpectedly, PMCA2 inhibition triggers rapid calcium-dependent loss of α7-nAChR clusters. This extreme regulatory response is mediated by CaMKII, involves proteasome activity, depends on the second intracellular loop of α7-nAChR subunits, and is specific in that it does not alter two other classes of calcium-permeable ionotropic receptors on the same neurons. A critical link is provided by the scaffold protein PSD-95 (postsynaptic density-95), which is associated with α7-nAChRs and constrains their mobility as revealed by single-particle tracking on neurons. The PSD-95 link is required for PMCA2-mediated removal of α7-nAChR clusters. This three-component combination of PMCA2, PSD-95, and α7-nAChR offers a novel mechanism for tight control of calcium dynamics in neurons.


Asunto(s)
Calcio/metabolismo , Interneuronas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/fisiología , Receptores Nicotínicos/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Homólogo 4 de la Proteína Discs Large , Femenino , Hipocampo/fisiología , Masculino , Péptidos/farmacología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/antagonistas & inhibidores , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7
16.
Proc Natl Acad Sci U S A ; 109(13): 4768-73, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22411830

RESUMEN

Chondroitin sulfate proteoglycans (CSPGs) represent a major barrier to regenerating axons in the central nervous system (CNS), but the structural diversity of their polysaccharides has hampered efforts to dissect the structure-activity relationships underlying their physiological activity. By taking advantage of our ability to chemically synthesize specific oligosaccharides, we demonstrate that a sugar epitope on CSPGs, chondroitin sulfate-E (CS-E), potently inhibits axon growth. Removal of the CS-E motif significantly attenuates the inhibitory activity of CSPGs on axon growth. Furthermore, CS-E functions as a protein recognition element to engage receptors including the transmembrane protein tyrosine phosphatase PTPσ, thereby triggering downstream pathways that inhibit axon growth. Finally, masking the CS-E motif using a CS-E-specific antibody reversed the inhibitory activity of CSPGs and stimulated axon regeneration in vivo. These results demonstrate that a specific sugar epitope within chondroitin sulfate polysaccharides can direct important physiological processes and provide new therapeutic strategies to regenerate axons after CNS injury.


Asunto(s)
Axones/patología , Axones/fisiología , Proteoglicanos Tipo Condroitín Sulfato/inmunología , Epítopos/inmunología , Regeneración Nerviosa/fisiología , Animales , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Axones/efectos de los fármacos , Conformación de Carbohidratos , Pollos , Proteoglicanos Tipo Condroitín Sulfato/química , Sulfatos de Condroitina/química , Sulfatos de Condroitina/inmunología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/metabolismo , Conos de Crecimiento/patología , Ratones , Neuritas/enzimología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Nat Chem Biol ; 8(3): 253-61, 2012 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-22267118

RESUMEN

The transcription factor cyclic AMP-response element binding protein (CREB) is a key regulator of many neuronal processes, including brain development, circadian rhythm and long-term memory. Studies of CREB have focused on its phosphorylation, although the diversity of CREB functions in the brain suggests additional forms of regulation. Here we expand on a chemoenzymatic strategy for quantifying glycosylation stoichiometries to characterize the functional roles of CREB glycosylation in neurons. We show that CREB is dynamically modified with an O-linked ß-N-acetyl-D-glucosamine sugar in response to neuronal activity and that glycosylation represses CREB-dependent transcription by impairing its association with CREB-regulated transcription coactivator (CRTC; also known as transducer of regulated CREB activity). Blocking glycosylation of CREB alters cellular function and behavioral plasticity, enhancing both axonal and dendritic growth and long-term memory consolidation. Our findings demonstrate a new role for O-glycosylation in memory formation and provide a mechanistic understanding of how glycosylation contributes to critical neuronal functions. Moreover, we identify a previously unknown mechanism for the regulation of activity-dependent gene expression, neural development and memory.


Asunto(s)
Acetilglucosamina/metabolismo , Proteína de Unión a CREB/metabolismo , Regulación de la Expresión Génica , Memoria a Largo Plazo , Animales , Proteína de Unión a CREB/química , Glicosilación , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo
19.
Nature ; 475(7357): 524-7, 2011 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-21796212

RESUMEN

Epstein-Barr virus-induced gene 2 (EBI2, also known as GPR183) is a G-protein-coupled receptor that is required for humoral immune responses; polymorphisms in the receptor have been associated with inflammatory autoimmune diseases. The natural ligand for EBI2 has been unknown. Here we describe the identification of 7α,25-dihydroxycholesterol (also called 7α,25-OHC or 5-cholesten-3ß,7α,25-triol) as a potent and selective agonist of EBI2. Functional activation of human EBI2 by 7α,25-OHC and closely related oxysterols was verified by monitoring second messenger readouts and saturable, high-affinity radioligand binding. Furthermore, we find that 7α,25-OHC and closely related oxysterols act as chemoattractants for immune cells expressing EBI2 by directing cell migration in vitro and in vivo. A critical enzyme required for the generation of 7α,25-OHC is cholesterol 25-hydroxylase (CH25H). Similar to EBI2 receptor knockout mice, mice deficient in CH25H fail to position activated B cells within the spleen to the outer follicle and mount a reduced plasma cell response after an immune challenge. This demonstrates that CH25H generates EBI2 biological activity in vivo and indicates that the EBI2-oxysterol signalling pathway has an important role in the adaptive immune response.


Asunto(s)
Hidroxicolesteroles/farmacología , Receptores de Superficie Celular/inmunología , Animales , Formación de Anticuerpos/inmunología , Linfocitos B , Línea Celular , Movimiento Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Hidroxicolesteroles/química , Hígado/química , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G , Ovinos , Linfocitos T/inmunología
20.
Anal Biochem ; 417(1): 103-11, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21704013

RESUMEN

Pathogenic Gram-negative bacteria are a major public health concern because they are causative agents of life-threatening hospital-acquired infections. Due to the increasing rates of resistance to available antibiotics, there is an urgent need to develop new drugs. Acetyl-coenzyme A carboxylase (ACCase) is a promising target for the development of novel antibiotics. We describe here the expression, purification, and enzymatic activity of recombinant ACCases from two clinically relevant Gram-negative pathogens, Acinetobacter baumannii and Klebsiella pneumoniae. Recombinant ACCase subunits (AccAD, AccB, and AccC) were expressed and purified, and the holoenzymes were reconstituted. ACCase enzyme activity was monitored by direct detection of malonyl-coenzyme A (malonyl-CoA) formation by liquid chromatography tandem mass spectrometry (LC-MS/MS). Steady-state kinetics experiments showed similar k(cat) and K(M) values for both enzymes. In addition, similar IC(50) values were observed for inhibition of both enzymes by a previously reported ACCase inhibitor. To provide a higher throughput assay suitable for inhibitor screening, we developed and validated a luminescence-based ACCase assay that monitors ATP depletion. Finally, we established an enzyme activity assay for the isolated AccAD (carboxyltransferase) subunit, which is useful for determining whether novel ACCase inhibitors inhibit the biotin carboxylase or carboxyltransferase site of ACCase. The methods described here could be applied toward the identification and characterization of novel inhibitors.


Asunto(s)
Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Acinetobacter baumannii/enzimología , Klebsiella pneumoniae/enzimología , Acetilcoenzima A/metabolismo , Adenosina Trifosfatasas/metabolismo , Biocatálisis , Ligasas de Carbono-Nitrógeno/metabolismo , Clonación Molecular , Fluorometría , Cinética , Malonil Coenzima A/metabolismo , Subunidades de Proteína/aislamiento & purificación , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA