Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Chem Senses ; 45(5): 333-346, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32333759

RESUMEN

Both canonical olfactory sensory neurons (OSNs) and sensory neurons belonging to the guanylate cyclase D (GCD) "necklace" subsystem are housed in the main olfactory epithelium, which is continuously bombarded by toxins, pathogens, and debris from the outside world. Canonical OSNs address this challenge, in part, by undergoing renewal through neurogenesis; however, it is not clear whether GCD OSNs also continuously regenerate and, if so, whether newborn GCD precursors follow a similar developmental trajectory to that taken by canonical OSNs. Here, we demonstrate that GCD OSNs are born throughout adulthood and can persist in the epithelium for several months. Phosphodiesterase 2A is upregulated early in the differentiation process, followed by the sequential downregulation of ß-tubulin and the upregulation of CART protein. The GCD and MS4A receptors that confer sensory responses upon GCD neurons are initially expressed midway through this process but become most highly expressed once CART levels are maximal late in GCD OSN development. GCD OSN maturation is accompanied by a horizontal migration of neurons toward the central, curved portions of the cul-de-sac regions where necklace cells are concentrated. These findings demonstrate that-like their canonical counterparts-GCD OSNs undergo continuous renewal and define a GCD-specific developmental trajectory linking neurogenesis, maturation, and migration.


Asunto(s)
Diferenciación Celular , Guanilato Ciclasa/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Regulación hacia Abajo , Femenino , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Neuronas Receptoras Olfatorias/citología , Tubulina (Proteína)/metabolismo , Regulación hacia Arriba
2.
Biosci Rep ; 39(1)2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30530863

RESUMEN

Turner syndrome (TS) is a congenital disease caused by complete or partial loss of one X chromosome. Low bone mineral status is a major phenotypic characteristic of TS that can not be fully explained by X chromosome loss, suggesting other autosomal-linked mutations may also exist. Therefore, the present study aimed to detect potential genetic mutations in TS through examination of copy number variation (CNV). Seventeen patients with TS and 15 healthy volunteer girls were recruited. Array-based comparative genomic hybridization (a-CGH) was performed on whole blood genomic DNA (gDMA) from the 17 TS patients and 15 healthy volunteer girls to identify potential CNVs. The abnormal CNV of one identified gene (CARD11) was verified by quantitative PCR. All cases diagnosed had TS based on genotype examination and physical characteristics, including short stature and premature ovarian failure. Three rare CNVs, located individually at 7p22.3, 7p22.2, and Xp22.33, where six genes (TTYH3, AMZ1, GNA12, BC038729, CARD11, and SHOX (stature homeobox)) are located, were found in TS patients. Quantitative PCR confirmed the CNV of CARD11 in the genome of TS patients. Our results indicate that CARD11 gene is one of the mutated genes involved in TS disease. However, this CNV is rare and its contribution to TS phenotype requires further study.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Cromosomas Humanos X/química , Variaciones en el Número de Copia de ADN , Guanilato Ciclasa/genética , Síndrome de Turner/genética , Adolescente , Antropometría , Proteínas Adaptadoras de Señalización CARD/deficiencia , Estudios de Casos y Controles , Niño , Preescolar , Hibridación Genómica Comparativa , Femenino , Genotipo , Guanilato Ciclasa/deficiencia , Humanos , Mutación , Fenotipo , Síndrome de Turner/diagnóstico , Síndrome de Turner/patología , Adulto Joven
3.
Biochem Biophys Res Commun ; 506(1): 48-52, 2018 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-30336982

RESUMEN

The MALT1 (Mucosa associated lymphoid tissue lymphoma translocation protein 1) paracaspase couples antigen receptors on lymphocytes to downstream signaling events. Activation of MALT1 is known to involve stimulus-dependent CBM complex formation, that is, the recruitment of BCL10-bound MALT1 to a CARD-Coiled Coil protein. Beyond this canonical, CBM-dependent mechanism of MALT1 activation, recent studies suggest that MALT1 protease activity may be triggered by alternative mechanisms. For instance, the E3-ligase TRAF6 can activate MALT1 proteolytic function and induce MALT1 auto-cleavage. However, the interplay between CBM and TRAF6 with regard to MALT1 activation has remained incompletely elucidated. Here, by generating CRISPR/Cas9-derived knock-out Jurkat T-cells, we show that TRAF6 was dispensable for CARD11/BCL10-dependent MALT1 activation upon T-cell stimulation. However, ectopically-expressed TRAF6 could induce MALT1 activity in Jurkat T-cells devoid of either CARD11 or BCL10. These data provide unequivocal evidence that TRAF6-mediated MALT1 activation does not require the upstream scaffold CARD11 or the interaction between MALT1 and BCL10. Thus, TRAF6 may be part of a previously unidentified non-canonical pathway that triggers MALT1 protease activity independently of canonical CBM signalosomes.


Asunto(s)
Proteína 10 de la LLC-Linfoma de Células B/genética , Proteínas Adaptadoras de Señalización CARD/genética , Guanilato Ciclasa/genética , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Factor 6 Asociado a Receptor de TNF/genética , Proteína 10 de la LLC-Linfoma de Células B/deficiencia , Proteínas Adaptadoras de Señalización CARD/deficiencia , Sistemas CRISPR-Cas , Activación Enzimática/efectos de los fármacos , Edición Génica/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Guanilato Ciclasa/deficiencia , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/metabolismo , Acetato de Tetradecanoilforbol/farmacología
4.
J Allergy Clin Immunol ; 141(5): 1818-1830.e2, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28826773

RESUMEN

BACKGROUND: Combined immunodeficiency (CID) is a T-cell defect frequently presenting with recurrent infections, as well as associated immune dysregulation manifesting as autoimmunity or allergic inflammation. OBJECTIVE: We sought to identify the genetic aberration in 4 related patients with CID, early-onset asthma, eczema, and food allergies, as well as autoimmunity. METHODS: We performed whole-exome sequencing, followed by Sanger confirmation, assessment of the genetic variant effect on cell signaling, and evaluation of the resultant immune function. RESULTS: A heterozygous novel c.C88T 1-bp substitution resulting in amino acid change R30W in caspase activation and recruitment domain family member 11 (CARD11) was identified by using whole-exome sequencing and segregated perfectly to family members with severe atopy only but was not found in healthy subjects. We demonstrate that the R30W mutation results in loss of function while also exerting a dominant negative effect on wild-type CARD11. The CARD11 defect altered the classical nuclear factor κB pathway, resulting in poor in vitro T-cell responses to mitogens and antigens caused by reduced secretion of IFN-γ and IL-2. CONCLUSION: Unlike patients with biallelic mutations in CARD11 causing severe CID, the R30W defect results in a less profound yet prominent susceptibility to infections, as well as multiorgan atopy and autoimmunity.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Guanilato Ciclasa/genética , Guanilato Ciclasa/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Adulto , Proteínas Adaptadoras de Señalización CARD/deficiencia , Preescolar , Femenino , Guanilato Ciclasa/deficiencia , Humanos , Interferón gamma/genética , Interleucina-2/genética , Masculino , Mutación , FN-kappa B/genética , Estudios Prospectivos , Estudios Retrospectivos , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Secuenciación del Exoma/métodos
5.
Invest Ophthalmol Vis Sci ; 58(11): 4826-4835, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973329

RESUMEN

Purpose: While nitric oxide (NO) donors are emerging as treatments for glaucoma, the mechanism by which NO lowers intraocular pressure (IOP) is unclear. NO activates the enzyme guanylyl cyclase (GC) to produce cyclic guanosine monophosphate. We studied the ocular effects of inhaled and topically applied NO gas in mice and lambs, respectively. Methods: IOP and aqueous humor (AqH) outflow were measured in WT and GC-1α subunit null (GC-1-/-) mice. Mice breathed 40 parts per million (ppm) NO in O2 or control gas (N2/O2). We also studied the effect of ocular NO gas exposure (80, 250, 500, and 1000 ppm) on IOP in anesthetized lambs. NO metabolites were measured in AqH and plasma. Results: In awake WT mice, breathing NO for 40 minutes lowered IOP from 14.4 ± 1.9 mm Hg to 10.9 ± 1.0 mm Hg (n = 11, P < 0.001). Comparable results were obtained in anesthetized WT mice (n = 10, P < 0.001). In awake or anesthetized GC-1-/- mice, IOP did not change under similar experimental conditions (P ≥ 0.08, n = 20). Breathing NO increased in vivo outflow facility in WT but not GC-1-/- mice (+13.7 ± 14.6% vs. -12.1 ± 9.4%, n = 4 each, P < 0.05). In lambs, ocular exposure to NO lowered IOP in a dose-dependent manner (-0.43 mm Hg/ppm NO; n = 5 with 40 total measurements; P = 0.04) without producing corneal pathology or altering pulmonary and systemic hemodynamics. After ocular NO exposure, NO metabolites were increased in AqH (n = 8, P < 0.001) but not in plasma. Conclusions: Breathing NO reduced IOP and increased outflow facility in a GC-dependent manner in mice. Exposure of ovine eyes to NO lowers IOP.


Asunto(s)
Humor Acuoso/fisiología , Guanilato Ciclasa/fisiología , Presión Intraocular/efectos de los fármacos , Óxido Nítrico/farmacología , Administración por Inhalación , Administración Tópica , Animales , Modelos Animales de Enfermedad , Femenino , Guanilato Ciclasa/deficiencia , Masculino , Ratones , Ratones Transgénicos , Óxido Nítrico/administración & dosificación , Ovinos
6.
Neuroscience ; 360: 180-189, 2017 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-28782641

RESUMEN

In hippocampus, two guanylyl cyclases (NO-GC1 and NO-GC2) are involved in the transduction of the effects of nitric oxide (NO) on synaptic transmission. However, the respective roles of the NO-GC isoforms on synaptic transmission are less clear in other regions of the brain. In the present study, we used knock-out mice deficient for the NO-GC1 isoform (NO-GC1 KO) to analyze its role in the glutamatergic and GABAergic neurotransmission at pyramidal neurons in layers II/III of somatosensory cortex. NO-GC1 KO slices revealed reduced frequencies of miniature excitatory- and inhibitory-postsynaptic currents, increased paired-pulse ratios and decreased input-output curves of evoked signals, which indicated the reduction of glutamate and GABA release in NO-GC1 KO mice. The functional changes in NO-GC1 KO mice were caused by the lack of cGMP as they were rescued to WT-like levels by the cGMP analog, 8-Br-PET-cGMP and conversely, mimicked by the NO-GC inhibitor, ODQ, in WT slices. In search of a cGMP target, two blockers of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (ZD7288 and DK-AH269) reduced glutamate release in WT to the level of NO-GC1 KO mice suggesting HCN channels as possible effectors for presynaptic cGMP enhancing the glutamate release probability. By blocking postsynaptic NMDA receptors, the NMDA receptor-dependent NO signal was shown to be linked to the effect of NO-GC1 on presynaptic GABA release. Of note, the balance between glutamatergic and GABAergic inputs at individual synapses remained unaltered in the NO-GC1 KO mice. In sum, our results indicate a role for cGMP generated by presynaptic localized NO-GC1 to adjust inhibitory and excitatory inputs at individual synapses in the somatosensory cortex.


Asunto(s)
GMP Cíclico/análogos & derivados , Ácido Glutámico/metabolismo , Guanilato Ciclasa/metabolismo , Óxido Nítrico/metabolismo , Receptores de Superficie Celular/metabolismo , Corteza Somatosensorial/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , GMP Cíclico/genética , GMP Cíclico/metabolismo , Guanilato Ciclasa/deficiencia , Hipocampo/metabolismo , Ratones Noqueados , Receptores de Superficie Celular/deficiencia , Guanilil Ciclasa Soluble/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
7.
Blood ; 126(14): 1658-69, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26289640

RESUMEN

Omenn syndrome (OS) is a severe immunodeficiency associated with erythroderma, lymphoproliferation, elevated IgE, and hyperactive oligoclonal T cells. A restricted T-cell repertoire caused by defective thymic T-cell development and selection, lymphopenia with homeostatic proliferation, and lack of regulatory T cells are considered key factors in OS pathogenesis. We report 2 siblings presenting with cytomegalovirus (CMV) and Pneumocystis jirovecii infections and recurrent sepsis; one developed all clinical features of OS. Both carried homozygous germline mutations in CARD11 (p.Cys150*), impairing NF-κB signaling and IL-2 production. A somatic second-site mutation reverting the stop codon to a missense mutation (p.Cys150Leu) was detected in tissue-infiltrating T cells of the OS patient. Expression of p.Cys150Leu in CARD11-deficient T cells largely reconstituted NF-κB signaling. The reversion likely occurred in a prethymic T-cell precursor, leading to a chimeric T-cell repertoire. We speculate that in our patient the functional advantage of the revertant T cells in the context of persistent CMV infection, combined with lack of regulatory T cells, may have been sufficient to favor OS. This first observation of OS in a patient with a T-cell activation defect suggests that severely defective T-cell development or homeostatic proliferation in a lymphopenic environment are not required for this severe immunopathology.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Guanilato Ciclasa/genética , Activación de Linfocitos/genética , Mutación , Inmunodeficiencia Combinada Grave/genética , Linfocitos T Reguladores/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/inmunología , Femenino , Citometría de Flujo , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/inmunología , Humanos , Immunoblotting , Inmunohistoquímica , Inmunofenotipificación , Lactante , Activación de Linfocitos/inmunología , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Inmunodeficiencia Combinada Grave/inmunología , Hermanos
8.
J Biol Chem ; 290(34): 20880-20892, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26124274

RESUMEN

Photoreceptor cyclic nucleotide-gated (CNG) channels play a pivotal role in phototransduction. Mutations in the cone CNG channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophies. We have shown endoplasmic reticulum (ER) stress-associated apoptotic cone death and increased phosphorylation of the ER Ca(2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in CNG channel-deficient mice. We also presented a remarkable elevation of cGMP and an increased activity of the cGMP-dependent protein kinase (protein kinase G, PKG) in CNG channel deficiency. This work investigated whether cGMP/PKG signaling regulates ER stress and IP3R1 phosphorylation in CNG channel-deficient cones. Treatment with PKG inhibitor and deletion of guanylate cyclase-1 (GC1), the enzyme producing cGMP in cones, were used to suppress cGMP/PKG signaling in cone-dominant Cnga3(-/-)/Nrl(-/-) mice. We found that treatment with PKG inhibitor or deletion of GC1 effectively reduced apoptotic cone death, increased expression levels of cone proteins, and decreased activation of Müller glial cells. Furthermore, we observed significantly increased phosphorylation of IP3R1 and reduced ER stress. Our findings demonstrate a role of cGMP/PKG signaling in ER stress and ER Ca(2+) channel regulation and provide insights into the mechanism of cone degeneration in CNG channel deficiency.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Estrés del Retículo Endoplásmico/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Animales , Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Carbazoles/farmacología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Proteínas del Ojo/genética , Regulación de la Expresión Génica , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones , Ratones Noqueados , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Células Fotorreceptoras Retinianas Conos/citología , Transducción de Señal , Tionucleótidos/farmacología
9.
Hypertension ; 65(2): 385-92, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25452469

RESUMEN

Nitroxyl (HNO), the reduced and protonated form of nitric oxide (NO·), confers unique physiological effects including vasorelaxation and enhanced cardiac contractility. These features have spawned current pharmaceutical development of HNO donors as heart failure therapeutics. HNO interacts with selective redox sensitive cysteines to effect signaling but is also proposed to activate soluble guanylate cyclase (sGC) in vitro to induce vasodilation and potentially enhance contractility. Here, we tested whether sGC stimulation is required for these HNO effects in vivo and if HNO also modifies a redox-sensitive cysteine (C42) in protein kinase G-1α to control vasorelaxation. Intact mice and isolated arteries lacking the sGC-ß subunit (sGCKO, results in full sGC deficiency) or expressing solely a redox-dead C42S mutant protein kinase G-1α were exposed to the pure HNO donor, CXL-1020. CXL-1020 induced dose-dependent systemic vasodilation while increasing contractility in controls; however, vasodilator effects were absent in sGCKO mice whereas contractility response remained. The CXL-1020 dose reversing 50% of preconstricted force in aortic rings was ≈400-fold greater in sGCKO than controls. Cyclic-GMP and cAMP levels were unaltered in myocardium exposed to CXL-1020, despite its inotropic-vasodilator activity. In protein kinase G-1α(C42S) mice, CXL-1020 induced identical vasorelaxation in vivo and in isolated aortic and mesenteric vessels as in littermate controls. In both groups, dilation was near fully blocked by pharmacologically inhibiting sGC. Thus, sGC and cGMP-dependent signaling are necessary and sufficient for HNO-induced vasodilation in vivo but are not required for positive inotropic action. Redox modulation of protein kinase G-1α is not a mechanism for HNO-mediated vasodilation.


Asunto(s)
Cardiotónicos/farmacología , Guanilato Ciclasa/fisiología , Óxidos de Nitrógeno/farmacología , Receptores Citoplasmáticos y Nucleares/fisiología , Vasodilatación/fisiología , Animales , Aorta/efectos de los fármacos , GMP Cíclico/fisiología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/química , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/deficiencia , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Cisteína/química , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/fisiopatología , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Óxido Nítrico/fisiología , Donantes de Óxido Nítrico/farmacología , Oxidación-Reducción , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Sistemas de Mensajero Secundario/fisiología , Guanilil Ciclasa Soluble , Sulfonamidas/farmacología , Vasodilatación/efectos de los fármacos
10.
Ann Rheum Dis ; 74(7): 1408-16, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24567525

RESUMEN

OBJECTIVES: We have previously described the antifibrotic role of the soluble guanylate cyclase (sGC). The mode of action, however, remained elusive. In the present study, we describe a novel link between sGC signalling and transforming growth factor ß (TGFß) signalling that mediates the antifibrotic effects of the sGC. METHODS: Human fibroblasts and murine sGC knockout fibroblasts were treated with the sGC stimulator BAY 41-2272 or the stable cyclic guanosine monophosphate (cGMP) analogue 8-Bromo-cGMP and stimulated with TGFß. sGC knockout fibroblasts were isolated from sGCI(fl/fl) mice, and recombination was induced by Cre-adenovirus. In vivo, we studied the antifibrotic effects of BAY 41-2272 in mice overexpressing a constitutively active TGF-ß1 receptor. RESULTS: sGC stimulation inhibited TGFß-dependent fibroblast activation and collagen release. sGC knockout fibroblasts confirmed that the sGC is essential for the antifibrotic effects of BAY 41-2272. Furthermore, 8-Bromo-cGMP reduced TGFß-dependent collagen release. While nuclear p-SMAD2 and 3 levels, SMAD reporter activity and transcription of classical TGFß target genes remained unchanged, sGC stimulation blocked the phosphorylation of ERK. In vivo, sGC stimulation inhibited TGFß-driven dermal fibrosis but did not change p-SMAD2 and 3 levels and TGFß target gene expression, confirming that non-canonical TGFß pathways mediate the antifibrotic sGC activity. CONCLUSIONS: We elucidated the antifibrotic mode of action of the sGC that increases cGMP levels, blocks non-canonical TGFß signalling and inhibits experimental fibrosis. Since sGC stimulators have shown excellent efficacy and tolerability in phase 3 clinical trials for pulmonary arterial hypertension, they may be further developed for the simultaneous treatment of fibrosis and vascular disease in systemic sclerosis.


Asunto(s)
Fibroblastos/patología , Guanilato Ciclasa/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/fisiopatología , Transducción de Señal/fisiología , Piel/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Estudios de Casos y Controles , Células Cultivadas , Colágeno/metabolismo , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis/metabolismo , Fibrosis/prevención & control , Guanilato Ciclasa/deficiencia , Humanos , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Pirazoles/farmacología , Piridinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal/efectos de los fármacos , Piel/metabolismo , Proteínas Smad/metabolismo , Guanilil Ciclasa Soluble , Factor de Crecimiento Transformador beta/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(9): 3602-7, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24550448

RESUMEN

Cone phototransduction and survival of cones in the human macula is essential for color vision and for visual acuity. Progressive cone degeneration in age-related macular degeneration, Stargardt disease, and recessive cone dystrophies is a major cause of blindness. Thyroid hormone (TH) signaling, which regulates cell proliferation, differentiation, and apoptosis, plays a central role in cone opsin expression and patterning in the retina. Here, we investigated whether TH signaling affects cone viability in inherited retinal degeneration mouse models. Retinol isomerase RPE65-deficient mice [a model of Leber congenital amaurosis (LCA) with rapid cone loss] and cone photoreceptor function loss type 1 mice (severe recessive achromatopsia) were used to determine whether suppressing TH signaling with antithyroid treatment reduces cone death. Further, cone cyclic nucleotide-gated channel B subunit-deficient mice (moderate achromatopsia) and guanylate cyclase 2e-deficient mice (LCA with slower cone loss) were used to determine whether triiodothyronine (T3) treatment (stimulating TH signaling) causes deterioration of cones. We found that cone density in retinol isomerase RPE65-deficient and cone photoreceptor function loss type 1 mice increased about sixfold following antithyroid treatment. Cone density in cone cyclic nucleotide-gated channel B subunit-deficient and guanylate cyclase 2e-deficient mice decreased about 40% following T3 treatment. The effect of TH signaling on cone viability appears to be independent of its regulation on cone opsin expression. This work demonstrates that suppressing TH signaling in retina dystrophy mouse models is protective of cones, providing insights into cone preservation and therapeutic interventions.


Asunto(s)
Defectos de la Visión Cromática/complicaciones , Amaurosis Congénita de Leber/complicaciones , Células Fotorreceptoras Retinianas Conos/fisiología , Degeneración Retiniana/prevención & control , Transducción de Señal/fisiología , Hormonas Tiroideas/metabolismo , Animales , Antitiroideos/farmacología , Defectos de la Visión Cromática/tratamiento farmacológico , Opsinas de los Conos/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Guanilato Ciclasa/deficiencia , Amaurosis Congénita de Leber/tratamiento farmacológico , Metimazol , Ratones , Ratones Noqueados , Receptores de Superficie Celular/deficiencia , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/etiología , Degeneración Retiniana/fisiopatología , Triyodotironina/farmacología , cis-trans-Isomerasas/deficiencia
12.
Nature ; 504(7480): 432-6, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24213632

RESUMEN

Myocardial infarction, a leading cause of death in the Western world, usually occurs when the fibrous cap overlying an atherosclerotic plaque in a coronary artery ruptures. The resulting exposure of blood to the atherosclerotic material then triggers thrombus formation, which occludes the artery. The importance of genetic predisposition to coronary artery disease and myocardial infarction is best documented by the predictive value of a positive family history. Next-generation sequencing in families with several affected individuals has revolutionized mutation identification. Here we report the segregation of two private, heterozygous mutations in two functionally related genes, GUCY1A3 (p.Leu163Phefs*24) and CCT7 (p.Ser525Leu), in an extended myocardial infarction family. GUCY1A3 encodes the α1 subunit of soluble guanylyl cyclase (α1-sGC), and CCT7 encodes CCTη, a member of the tailless complex polypeptide 1 ring complex, which, among other functions, stabilizes soluble guanylyl cyclase. After stimulation with nitric oxide, soluble guanylyl cyclase generates cGMP, which induces vasodilation and inhibits platelet activation. We demonstrate in vitro that mutations in both GUCY1A3 and CCT7 severely reduce α1-sGC as well as ß1-sGC protein content, and impair soluble guanylyl cyclase activity. Moreover, platelets from digenic mutation carriers contained less soluble guanylyl cyclase protein and consequently displayed reduced nitric-oxide-induced cGMP formation. Mice deficient in α1-sGC protein displayed accelerated thrombus formation in the microcirculation after local trauma. Starting with a severely affected family, we have identified a link between impaired soluble-guanylyl-cyclase-dependent nitric oxide signalling and myocardial infarction risk, possibly through accelerated thrombus formation. Reversing this defect may provide a new therapeutic target for reducing the risk of myocardial infarction.


Asunto(s)
Susceptibilidad a Enfermedades/metabolismo , Infarto del Miocardio/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Chaperonina con TCP-1/genética , Chaperonina con TCP-1/metabolismo , GMP Cíclico/metabolismo , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Mutación/genética , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Linaje , Activación Plaquetaria , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Reproducibilidad de los Resultados , Solubilidad , Guanilil Ciclasa Soluble , Trombosis/metabolismo , Vasodilatación
13.
Chem Senses ; 38(9): 769-81, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24122319

RESUMEN

The main olfactory system of mice contains a small subset of olfactory sensory neurons (OSNs) that are stimulated by CO2. The objective of this study was to record olfactory receptor responses to a range of CO2 concentrations to further elucidate steps in the proposed CO2 transduction pathway in mice. Electro-olfactograms (EOGs) were recorded before and after inhibiting specific steps in the CO2 transduction pathway with topically applied inhibitors. Inhibition of extracellular carbonic anhydrase (CA) did not significantly affect EOG responses to CO2 but did decrease EOG responses to several control odorants. Inhibition of intracellular CA or cyclic nucleotide-gated channels attenuated EOG responses to CO2, confirming the role of these components in CO2 sensing in mice. We also show that, like canonical OSNs, CO2-sensitive OSNs depend on Ca²âº-activated Cl⁻ channels for depolarization of receptor neurons. Lastly, we found that guanylyl cyclase-D knockout mice were still able to respond to CO2, indicating that other pathways may exist for the detection of low concentrations of nasal CO2. We discuss these findings as they relate to previous studies on CO2-sensitive OSNs in mice and other animals.


Asunto(s)
Dióxido de Carbono/farmacología , Guanilato Ciclasa/genética , Mucosa Nasal/efectos de los fármacos , Receptores de Superficie Celular/genética , Animales , Canales de Cloruro/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Fenómenos Electrofisiológicos , Femenino , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Nasal/fisiología , Neuronas Receptoras Olfatorias/metabolismo , Pentanoles/farmacología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Olfato/fisiología
14.
J Neurosci ; 33(37): 14939-48, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24027293

RESUMEN

Photoreceptor cyclic nucleotide-gated (CNG) channels regulate Ca(2+) influx in rod and cone photoreceptors. cGMP, the native ligand of the photoreceptor CNG channels, has been associated with cytotoxicity when its levels rise above normal due to defects in photoreceptor phosphodiesterase (PDE6) or regulation of retinal guanylyl cyclase (retGC). We found a massive accumulation of cGMP in CNGA3-deficient retina and investigated whether cGMP accumulation plays a role in cone degeneration in CNG channel deficiency. The time course study showed that the retinal cGMP level in Cnga3(-/-);Nrl(-/-) mice with CNGA3 deficiency on a cone-dominant background was sharply increased at postnatal day 8 (P8), peaked around P10-P15, remained high through P30-P60, and returned to near control level at P90. This elevation pattern correlated with photoreceptor apoptotic death, which peaked around P15-P20. In Cnga3(-/-);Gucy2e(-/-) mice lacking retGC1, cone density and expression levels of cone-specific proteins were significantly increased compared with Cnga3(-/-), consistent with a role of cGMP accumulation as the major contributor to cone death caused by CNG channel deficiency. The activity and expression levels of cGMP-dependent protein kinase G (PKG) were significantly increased in Cnga3(-/-);Nrl(-/-) retina compared with Nrl(-/-), suggesting an involvement of PKG regulation in cell death. Our results indicate that cGMP accumulation in photoreceptors can itself exert cytotoxic effect in cones, independently of CNG channel activity and Ca(2+) influx.


Asunto(s)
GMP Cíclico/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Animales , Animales Recién Nacidos , Proteínas Portadoras , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo/metabolismo , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/metabolismo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Receptores de N-Metil-D-Aspartato , Retina/patología
15.
Am J Respir Crit Care Med ; 188(7): 789-99, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23841447

RESUMEN

RATIONALE: Soluble guanylyl cyclase (sGC), a cyclic guanosine 5'-monophosphate-generating enzyme, regulates smooth muscle tone and exerts antiinflammatory effects in animal models of asthma and acute lung injury. In chronic obstructive pulmonary disease (COPD), primarily caused by cigarette smoke (CS), lung inflammation persists and smooth muscle tone remains elevated, despite ample amounts of nitric oxide that could activate sGC. OBJECTIVES: To determine the expression and function of sGC in patients with COPD and in a murine model of COPD. METHODS: Expression of sGCα1, α2, and ß1 subunits was examined in lungs of never-smokers, smokers without airflow limitation, and patients with COPD; and in C57BL/6 mice after 3 days, 4 weeks, and 24 weeks of CS exposure. The functional role of sGC was investigated in vivo by measuring bronchial responsiveness to serotonin in mice using genetic and pharmacologic approaches. MEASUREMENTS AND MAIN RESULTS: Pulmonary expression of sGC, both at mRNA and protein level, was decreased in smokers without airflow limitation and in patients with COPD, and correlated with disease severity (FEV1%). In mice, exposure to CS reduced sGC, cyclic guanosine 5'-monophosphate levels, and protein kinase G activity. sGCα1(-/-) mice exposed to CS exhibited bronchial hyperresponsiveness to serotonin. Activation of sGC by BAY 58-2667 restored the sGC signaling and attenuated bronchial hyperresponsiveness in CS-exposed mice. CONCLUSIONS: Down-regulation of sGC because of CS exposure might contribute to airflow limitation in COPD.


Asunto(s)
Guanilato Ciclasa/fisiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Receptores Citoplasmáticos y Nucleares/fisiología , Serotonina/fisiología , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Anciano , Animales , Broncoconstricción/efectos de los fármacos , Broncoconstricción/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Guanilato Ciclasa/análisis , Guanilato Ciclasa/deficiencia , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptores Citoplasmáticos y Nucleares/análisis , Receptores Citoplasmáticos y Nucleares/deficiencia , Mucosa Respiratoria/química , Fumar/fisiopatología , Guanilil Ciclasa Soluble
16.
Chem Senses ; 38(5): 391-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23564012

RESUMEN

Rodents rely on olfactory stimuli to communicate information between conspecifics that is critical for health and survival. For example, rodents that detect a food odor simultaneously with the social odor carbon disulfide (CS(2)) will acquire a preference for that food. Disruption of the chemosensory transduction cascade in CS(2-)sensitive olfactory sensory neurons (OSNs) that express the receptor guanylyl cyclase type D (GC-D; GC-D+ OSNs) will prevent mice from acquiring these preferences. GC-D+ OSNs also respond to the natriuretic peptide uroguanylin, which is excreted into urine and feces. We analyzed if uroguanylin could also act as a social stimulus to promote the acquisition of food preferences. We found that feces of mice that had eaten odored food, but not unodored food, promoted a strong preference for that food in mice exposed to the feces. Olfactory exploration of uroguanylin presented with a food odor similarly produced a preference that was absent when mice were exposed to the food odor alone. Finally, the acquisition of this preference was dependent on GC-D+ OSNs, as mice lacking GC-D (Gucy2d(-)(/-) mice) showed no preference for the demonstrated food. Together with our previous findings, these results demonstrate that the diverse activators of GC-D+ OSNs elicit a common behavioral result and suggest that this specialized olfactory subsystem acts as a labeled line for a type of associative olfactory learning.


Asunto(s)
Preferencias Alimentarias/efectos de los fármacos , Guanilato Ciclasa/metabolismo , Péptidos Natriuréticos/farmacología , Neuronas Receptoras Olfatorias/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Animales , Guanilato Ciclasa/deficiencia , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Receptoras Olfatorias/enzimología , Neuronas Receptoras Olfatorias/metabolismo , Receptores de Superficie Celular/deficiencia
17.
J Allergy Clin Immunol ; 131(5): 1376-83.e3, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23561803

RESUMEN

BACKGROUND: Primary immunodeficiencies represent model diseases for the mechanistic understanding of the human innate and adaptive immune response. They are clinically highly relevant per se because in patients with severe combined immunodeficiency (SCID), infections caused by opportunistic pathogens are typically life-threatening early in life. OBJECTIVES: We aimed at defining and functionally characterizing a novel form of SCID in an infant of consanguineous parents who presented with life-threatening Pneumocystis jirovecii pneumonia using a comprehensive immunologic and whole-exome genetic diagnostic strategy. METHODS: Analysis of leukocyte subpopulations was performed by using multicolor flow cytometry and was combined with stimulation tests for T-cell function. The search for a disease-causing mutation was performed with diagnostic whole-exome sequencing and systematic variant categorization. Reconstitution assays were used for validating the loss-of-function mutation. RESULTS: The novel entity of SCID was characterized by agammaglobulinemia and profoundly deficient T-cell function despite quantitatively normal T and B lymphocytes. Genetic analysis revealed a single pathogenic homozygous nonsense mutation of the caspase recruitment domain 11 (CARD11) gene. In reconstitution assays we demonstrated that the patient-derived truncated CARD11 protein is defective in antigen receptor signaling and nuclear factor κB activation. CONCLUSION: We show that an inactivating CARD11 mutation links defective nuclear factor κB signaling to a novel cause of autosomal recessive SCID.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/genética , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/genética , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Secuencia de Aminoácidos , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Línea Celular , Codón sin Sentido , Femenino , Guanilato Ciclasa/antagonistas & inhibidores , Homocigoto , Humanos , Lactante , Células Jurkat , Datos de Secuencia Molecular , Linaje , Inmunodeficiencia Combinada Grave/complicaciones
18.
PLoS One ; 8(3): e60156, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527308

RESUMEN

Primary open angle glaucoma (POAG) is a leading cause of blindness worldwide. The molecular signaling involved in the pathogenesis of POAG remains unknown. Here, we report that mice lacking the α1 subunit of the nitric oxide receptor soluble guanylate cyclase represent a novel and translatable animal model of POAG, characterized by thinning of the retinal nerve fiber layer and loss of optic nerve axons in the context of an open iridocorneal angle. The optic neuropathy associated with soluble guanylate cyclase α1-deficiency was accompanied by modestly increased intraocular pressure and retinal vascular dysfunction. Moreover, data from a candidate gene association study suggests that a variant in the locus containing the genes encoding for the α1 and ß1 subunits of soluble guanylate cyclase is associated with POAG in patients presenting with initial paracentral vision loss, a disease subtype thought to be associated with vascular dysregulation. These findings provide new insights into the pathogenesis and genetics of POAG and suggest new therapeutic strategies for POAG.


Asunto(s)
Modelos Animales de Enfermedad , Glaucoma de Ángulo Abierto/enzimología , Glaucoma de Ángulo Abierto/fisiopatología , Guanilato Ciclasa/deficiencia , Nervio Óptico/patología , Receptores Citoplasmáticos y Nucleares/deficiencia , Neuronas Retinianas/patología , Análisis de Varianza , Animales , Femenino , Guanilato Ciclasa/genética , Inmunohistoquímica , Presión Intraocular/fisiología , Ratones , Ratones Noqueados , Ratones Mutantes , Oftalmoscopía , Fenilendiaminas , Receptores Citoplasmáticos y Nucleares/genética , Guanilil Ciclasa Soluble , Tomografía de Coherencia Óptica
19.
J Allergy Clin Immunol ; 131(2): 477-85.e1, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23374270

RESUMEN

BACKGROUND: Profound combined immunodeficiency can present with normal numbers of T and B cells, and therefore the functional defect of the cellular and humoral immune response is often not recognized until the first severe clinical manifestation. Here we report a patient of consanguineous descent presenting at 13 months of age with hypogammaglobulinemia, Pneumocystis jirovecii pneumonia, and a suggestive family history. OBJECTIVE: We sought to identify the genetic alteration in a patient with combined immunodeficiency and characterize human caspase recruitment domain family, member 11 (CARD11), deficiency. METHODS: Molecular, immunologic, and functional assays were performed. RESULTS: The immunologic characterization revealed only subtle changes in the T-cell and natural killer cell compartment, whereas B-cell differentiation, although normal in number, was distinctively blocked at the transitional stage. Genetic evaluation revealed a homozygous deletion of exon 21 in CARD11 as the underlying defect. This deletion abrogated protein expression and activation of the canonical nuclear factor κB (NF-κB) pathway in lymphocytes after antigen receptor or phorbol 12-myristate 13-acetate stimulation, whereas CD40 signaling in B cells was preserved. The abrogated activation of the canonical NF-κB pathway was associated with severely impaired upregulation of inducible T-cell costimulator, OX40, cytokine production, proliferation of T cells, and B cell-activating factor receptor expression on B cells. CONCLUSION: Thus in patients with CARD11 deficiency, the combination of impaired activation and especially upregulation of inducible T-cell costimulator on T cells, together with severely disturbed peripheral B-cell differentiation, apparently leads to a defective T-cell/B-cell cooperation and probably germinal center formation and clinically results in severe immunodeficiency. This report discloses the crucial and nonredundant role of canonical NF-κB activation and specifically CARD11 in the antigen-specific immune response in human subjects.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/deficiencia , Guanilato Ciclasa/deficiencia , Síndromes de Inmunodeficiencia/enzimología , Eliminación de Secuencia , Agammaglobulinemia/enzimología , Agammaglobulinemia/genética , Agammaglobulinemia/inmunología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Femenino , Guanilato Ciclasa/genética , Guanilato Ciclasa/inmunología , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Lactante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA