Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell ; 186(26): 5812-5825.e21, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38056462

RESUMEN

Acyl-coenzyme A (acyl-CoA) species are cofactors for numerous enzymes that acylate thousands of proteins. Here, we describe an enzyme that uses S-nitroso-CoA (SNO-CoA) as its cofactor to S-nitrosylate multiple proteins (SNO-CoA-assisted nitrosylase, SCAN). Separate domains in SCAN mediate SNO-CoA and substrate binding, allowing SCAN to selectively catalyze SNO transfer from SNO-CoA to SCAN to multiple protein targets, including the insulin receptor (INSR) and insulin receptor substrate 1 (IRS1). Insulin-stimulated S-nitrosylation of INSR/IRS1 by SCAN reduces insulin signaling physiologically, whereas increased SCAN activity in obesity causes INSR/IRS1 hypernitrosylation and insulin resistance. SCAN-deficient mice are thus protected from diabetes. In human skeletal muscle and adipose tissue, SCAN expression increases with body mass index and correlates with INSR S-nitrosylation. S-nitrosylation by SCAN/SNO-CoA thus defines a new enzyme class, a unique mode of receptor tyrosine kinase regulation, and a revised paradigm for NO function in physiology and disease.


Asunto(s)
Insulina , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Transducción de Señal , Animales , Humanos , Ratones , Acilcoenzima A/metabolismo , Tejido Adiposo/metabolismo , Resistencia a la Insulina , Óxido Nítrico/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo
2.
Antioxid Redox Signal ; 39(10-12): 621-634, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37053107

RESUMEN

Aims: S-nitrosylation of proteins is the main mechanism through which nitric oxide (NO) regulates cellular function and likely represents the archetype redox-based signaling system across aerobic and anaerobic organisms. How NO generated by different nitric oxide synthase (NOS) isoforms leads to specificity of S-nitrosylation remains incompletely understood. This study aimed to identify proteins interacting with, and whose S-nitrosylation is mediated by, human NOS isoforms in the same cellular system, thereby illuminating the contribution of individual NOSs to specificity. Results: Of the hundreds of proteins interacting with each NOS, many were also S-nitrosylated. However, a large proportion of S-nitrosylated proteins (SNO-proteins) did not associate with NOS. Moreover, most NOS interactors and SNO-proteins were unique to each isoform. The amount of NO produced by each NOS isoform was unrelated to the numbers of SNO-proteins. Thus, NOSs promoted S-nitrosylation of largely distinct sets of target proteins. Different signaling pathways were enriched downstream of each NOS. Innovation and Conclusion: The interactomes and SNOomes of individual NOS isoforms were largely distinct. Only a small fraction of SNO-proteins interacted with their respective NOS. Amounts of S-nitrosylation were unrelated to the amount of NO generated by NOSs. These data argue against free diffusion of NO or NOS interactions as being necessary or sufficient for S-nitrosylation and favor roles for additional enzymes and/or regulatory elements in imparting SNO-protein specificity. Antioxid. Redox Signal. 39, 621-634.


Asunto(s)
Óxido Nítrico Sintasa , Proteoma , Humanos , Proteoma/metabolismo , Óxido Nítrico Sintasa/metabolismo , Oxidación-Reducción , Transducción de Señal , Óxido Nítrico/metabolismo , Isoformas de Proteínas/metabolismo
3.
Mol Cell ; 82(16): 3089-3102.e7, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35931084

RESUMEN

The ß2-adrenergic receptor (ß2AR), a prototypic G-protein-coupled receptor (GPCR), is a powerful driver of bronchorelaxation, but the effectiveness of ß-agonist drugs in asthma is limited by desensitization and tachyphylaxis. We find that during activation, the ß2AR is modified by S-nitrosylation, which is essential for both classic desensitization by PKA as well as desensitization of NO-based signaling that mediates bronchorelaxation. Strikingly, S-nitrosylation alone can drive ß2AR internalization in the absence of traditional agonist. Mutant ß2AR refractory to S-nitrosylation (Cys265Ser) exhibits reduced desensitization and internalization, thereby amplifying NO-based signaling, and mice with Cys265Ser mutation are resistant to bronchoconstriction, inflammation, and the development of asthma. S-nitrosylation is thus a central mechanism in ß2AR signaling that may be operative widely among GPCRs and targeted for therapeutic gain.


Asunto(s)
Asma , Animales , Asma/inducido químicamente , Asma/genética , Ratones , Transducción de Señal
4.
Matrix Biol ; 28(7): 406-15, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19520159

RESUMEN

The adult olfactory epithelium has maintained the ability to reconstitute its olfactory sensory neurons (OSNs) from a basal progenitor cell compartment. This allows for life-long turnover and replacement of receptor components as well as repair of the primary olfactory pathway in response to injury and environmental insults. The present study investigated whether fibulin-3, a glycoprotein in the extracellular matrix and binding partner of tissue inhibitor of metalloproteinases-3 (TIMP-3), plays a role in ongoing plasticity and regenerative events in the adult primary olfactory pathway. In wild-type control mice, fibulin-3 protein was detected on IB4(+)CD31(+) blood vessels, nerve fascicles and the basement membrane underneath the olfactory epithelium. After target ablation (olfactory bulbectomy), fibulin-3 was also abundantly present in the central nervous system (CNS) scar tissue that occupied the bulbar cavity. Using two different lesion models, i.e. intranasal Triton X-100 lesion and olfactory bulbectomy, we show that fibulin-3 deficient (Efemp1(-/-)) mice have impaired recovery of the olfactory epithelium after injury. Ten days post-injury, Efemp1(-/-) mice showed altered basal stem/progenitor cell proliferation and increased overall numbers of mature (olfactory marker protein (OMP) -positive) versus immature OSNs. However, compromised regenerative capacity of the primary olfactory pathway in Efemp1(-/-) mice was evidenced by reduced numbers of mature OSNs at the later time point of 42 days post-injury. In addition to these neural differences there were consistent changes in blood vessel structure in the olfactory lamina propria of Efemp1(-/-) mice. Overall, these data suggest a role for fibulin-3 in tissue maintenance and regeneration in the adult olfactory pathway.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Regeneración Nerviosa/fisiología , Vías Olfatorias/fisiología , Animales , Proteínas de la Matriz Extracelular/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Bulbo Olfatorio/citología , Bulbo Olfatorio/patología , Bulbo Olfatorio/fisiología , Mucosa Olfatoria/citología , Mucosa Olfatoria/patología , Mucosa Olfatoria/fisiología , Vías Olfatorias/anatomía & histología , Vías Olfatorias/patología , Células Madre/citología , Células Madre/fisiología
5.
Hum Mol Genet ; 16(24): 3059-70, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17872905

RESUMEN

A mutation in the EFEMP1 gene causes Malattia Leventinese, an inherited macular degenerative disease with strong similarities to age-related macular degeneration. EFEMP1 encodes fibulin-3, an extracellular matrix protein of unknown function. To investigate its biological role, the murine Efemp1 gene was inactivated through targeted disruption. Efemp1(-/-) mice exhibited reduced reproductivity, and displayed an early onset of aging-associated phenotypes including reduced lifespan, decreased body mass, lordokyphosis, reduced hair growth, and generalized fat, muscle and organ atrophy. However, these mice appeared to have normal wound healing ability. Efemp1(-/-) mice on a C57BL/6 genetic background developed multiple large hernias including inguinal hernias, pelvic prolapse and protrusions of the xiphoid process. In contrast, Efemp1(-/-) mice on a BALB/c background rarely had any forms of hernias, indicating the presence of modifiers for fibulin-3's function in different mouse strains. Histological analysis revealed a marked reduction of elastic fibers in fascia, a thin layer of connective tissue maintaining and protecting structures throughout the body. No apparent macular degeneration associated defects were found in Efemp1(-/-) mice, suggesting that loss of fibulin-3 function is not the mechanism by which the mutation in EFEMP1 causes macular degeneration. These data demonstrate that fibulin-3 plays an important role in maintaining the integrity of fascia connective tissues and regulates aging.


Asunto(s)
Envejecimiento Prematuro/genética , Proteínas de la Matriz Extracelular/genética , Hernia/genética , Envejecimiento Prematuro/patología , Animales , Tejido Elástico/metabolismo , Tejido Elástico/patología , Proteínas de la Matriz Extracelular/fisiología , Fascia/metabolismo , Fascia/patología , Hernia/patología , Longevidad/genética , Degeneración Macular/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Reproducción/fisiología , Cicatrización de Heridas/fisiología
6.
Hum Mol Genet ; 16(20): 2423-32, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17664227

RESUMEN

Malattia leventinese (ML) is a dominantly inherited macular degenerative disease characterized by the presence of sub-retinal pigment epithelium (RPE) deposits. With the exception of an earlier age of onset, ML patients exhibit symptoms and histopathology compatible with the diagnosis of age-related macular degeneration (AMD), the most common cause of incurable blindness. ML is caused by a mutation (R345W) in the gene EFEMP1 which encodes fibulin-3, a protein of unknown function. We generated a knock-in mouse carrying the disease-associated mutation in the murine Efemp1 gene. Small, isolated sub-RPE deposits developed as early as 4 months of age in both heterozygous and homozygous knock-in mice. Over time these deposits increased in size and number eventually becoming continuous sheets. In older mice membranous debris was observed within the deposits and within Bruch's membrane, and was accompanied by general RPE and choroidal abnormalities including degeneration, vacuolation, loss or disruption of the RPE basal infoldings, choroidal atrophy, and focal thickening of and invasion of cellular processes into Bruch's membrane. Fibulin-3 was found to accumulate in the sub-RPE deposits. Thus, the Efemp1 knock-in mice reconstitute the most important histopathologic symptoms of both ML and AMD. We conclude that these mice are a valuable tool for studying the primary pathogenic course of basal deposits associated with macular degeneration and for testing prevention and treatment strategies for this class of diseases.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Degeneración Macular/genética , Degeneración Macular/patología , Epitelio Pigmentado Ocular/patología , Mutación Puntual , Animales , Arginina/genética , Lámina Basal de la Coroides/patología , Lámina Basal de la Coroides/ultraestructura , Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Nervio Óptico/patología , Epitelio Pigmentado Ocular/metabolismo , Epitelio Pigmentado Ocular/ultraestructura , Retina/patología , Factores de Tiempo , Triptófano/genética
7.
Mol Cell Biol ; 26(5): 1700-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16478991

RESUMEN

Elastic fibers provide tissues with elasticity which is critical to the function of arteries, lungs, skin, and other dynamic organs. Loss of elasticity is a major contributing factor in aging and diseases. However, the mechanism of elastic fiber development and assembly is poorly understood. Here, we show that lack of fibulin-4, an extracellular matrix molecule, abolishes elastogenesis. fibulin-4-/- mice generated by gene targeting exhibited severe lung and vascular defects including emphysema, artery tortuosity, irregularity, aneurysm, rupture, and resulting hemorrhages. All the homozygous mice died perinatally. The earliest abnormality noted was a uniformly narrowing of the descending aorta in fibulin-4-/- embryos at embryonic day 12.5 (E12.5). Aorta tortuosity and irregularity became noticeable at E15.5. Histological analysis demonstrated that fibulin-4-/- mice do not develop intact elastic fibers but contain irregular elastin aggregates. Electron microscopy revealed that the elastin aggregates are highly unusual in that they contain evenly distributed rod-like filaments, in contrast to the amorphous appearance of normal elastic fibers. Desmosine analysis indicated that elastin cross-links in fibulin-4-/- tissues were largely diminished. However, expression of tropoelastin or lysyl oxidase mRNA was unaffected in fibulin-4-/- mice. In addition, fibulin-4 strongly interacts with tropoelastin and colocalizes with elastic fibers in culture. These results demonstrate that fibulin-4 plays an irreplaceable role in elastogenesis.


Asunto(s)
Tejido Elástico/fisiología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Muerte Fetal/genética , Animales , Aorta/anomalías , Aorta/embriología , Células Cultivadas , Desmosina/metabolismo , Tejido Elástico/anomalías , Elastina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Silenciador del Gen , Humanos , Pulmón/anomalías , Pulmón/embriología , Pulmón/patología , Ratones , Ratones Mutantes , Proteína-Lisina 6-Oxidasa/metabolismo , Tropoelastina/metabolismo
8.
Proc Natl Acad Sci U S A ; 99(20): 13067-72, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12242346

RESUMEN

Malattia Leventinese (ML), an inherited macular degenerative disease, is closely reminiscent of age-related macular degeneration (AMD), the most common cause of incurable blindness. Both ML and AMD are characterized by extracellular deposits known as drusen between the retinal pigment epithelium (RPE) and Bruch's membrane. The mechanism underlying drusen formation is unknown. An Arg to Trp mutation in a gene of unknown function, EFEMP1, is responsible for ML, indicating EFEMP1 may be important in drusen formation. Here, we show that wild-type EFEMP1 is a secreted protein whereas mutant EFEMP1 is misfolded, secreted inefficiently, and retained within cells. In normal eyes, EFEMP1 is not present at the site of drusen formation. However, in ML eyes, EFEMP1 accumulates within the RPE cells and between the RPE and drusen, but does not appear to be a major component of drusen. Furthermore, in AMD eyes, EFEMP1 is found to accumulate beneath the RPE immediately overlaying drusen, but not in the region where there is no apparent retinal pathology observed. These data present evidence that misfolding and aberrant accumulation of EFEMP1 may cause drusen formation and cellular degeneration and play an important role in the etiology of both ML and AMD.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Degeneración Macular/metabolismo , Factores de Edad , Anciano , Anciano de 80 o más Años , Anticuerpos/metabolismo , Línea Celular , ADN Complementario/metabolismo , Femenino , Biblioteca de Genes , Glutatión Transferasa/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Degeneración Macular/genética , Degeneración Macular/patología , Mutación , Epitelio Pigmentado Ocular/metabolismo , Pruebas de Precipitina , Pliegue de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Retina/patología , Drusas Retinianas/metabolismo , Factores de Tiempo , Transfección
9.
J Biol Chem ; 277(34): 30591-7, 2002 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-12058047

RESUMEN

Bestrophin is a 68-kDa basolateral plasma membrane protein expressed in retinal pigment epithelial cells (RPE). It is encoded by the VMD2 gene, which is mutated in Best macular dystrophy, a disease characterized by a depressed light peak in the electrooculogram. Recently it was proposed that bestrophin is a chloride channel responsible for generating the light peak. To investigate its function further, we immunoaffinity purified a bestrophin complex from RPE lysates and identified bestrophin and the beta-catalytic subunit of protein phosphatase 2A (PP2A) as members of the complex by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Protein-protein interaction between bestrophin and PP2Ac and the structural subunit of PP2A, PR65, was confirmed by reciprocal immunoprecipitation. The C-terminal cytoplasmic domain of bestrophin was sufficient for the interaction with PP2A as demonstrated by a pulldown assay using a fusion of this domain with glutathione S-transferase. Bestrophin was phosphorylated when expressed in RPE-J cells and this phosphorylation was sensitive to okadaic acid. Purified PP2A effectively dephosphorylated bestrophin in vitro. These data suggest that bestrophin is in the signal transduction pathway that modulates the light peak of the electrooculogram, that it is regulated by phosphorylation, and that phosphorylation of bestrophin is in turn regulated by PP2A.


Asunto(s)
Proteínas del Ojo/fisiología , Fosfoproteínas Fosfatasas/fisiología , Secuencia de Aminoácidos , Animales , Bestrofinas , Células Cultivadas , Canales de Cloruro , Citosol/química , Electrooculografía , Proteínas del Ojo/química , Humanos , Luz , Datos de Secuencia Molecular , Fosfoproteínas Fosfatasas/química , Fosforilación , Proteína Fosfatasa 2 , Transducción de Señal , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA