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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(26): 13006-13015, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31189595

RESUMEN

Abdominal aortic aneurysm (AAA) remains the second most frequent vascular disease with high mortality but has no approved medical therapy. We investigated the direct role of apelin (APLN) in AAA and identified a unique approach to enhance APLN action as a therapeutic intervention for this disease. Loss of APLN potentiated angiotensin II (Ang II)-induced AAA formation, aortic rupture, and reduced survival. Formation of AAA was driven by increased smooth muscle cell (SMC) apoptosis and oxidative stress in Apln-/y aorta and in APLN-deficient cultured murine and human aortic SMCs. Ang II-induced myogenic response and hypertension were greater in Apln-/y mice, however, an equivalent hypertension induced by phenylephrine, an α-adrenergic agonist, did not cause AAA or rupture in Apln-/y mice. We further identified Ang converting enzyme 2 (ACE2), the major negative regulator of the renin-Ang system (RAS), as an important target of APLN action in the vasculature. Using a combination of genetic, pharmacological, and modeling approaches, we identified neutral endopeptidase (NEP) that is up-regulated in human AAA tissue as a major enzyme that metabolizes and inactivates APLN-17 peptide. We designed and synthesized a potent APLN-17 analog, APLN-NMeLeu9-A2, that is resistant to NEP cleavage. This stable APLN analog ameliorated Ang II-mediated adverse aortic remodeling and AAA formation in an established model of AAA, high-fat diet (HFD) in Ldlr-/- mice. Our findings define a critical role of APLN in AAA formation through induction of ACE2 and protection of vascular SMCs, whereas stable APLN analogs provide an effective therapy for vascular diseases.


Asunto(s)
Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/patología , Apelina/metabolismo , Neprilisina/metabolismo , Anciano , Anciano de 80 o más Años , Angiotensina II/administración & dosificación , Enzima Convertidora de Angiotensina 2 , Animales , Aorta Abdominal/citología , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Aneurisma de la Aorta Abdominal/etiología , Apelina/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/farmacología , Fármacos Cardiovasculares/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Miocitos del Músculo Liso , Neprilisina/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Peptidil-Dipeptidasa A/metabolismo , Fenilefrina/administración & dosificación , Cultivo Primario de Células , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética
2.
Circulation ; 137(20): 2152-2165, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29348261

RESUMEN

BACKGROUND: Defining conserved molecular pathways in animal models of successful cardiac regeneration could yield insight into why adult mammals have inadequate cardiac regeneration after injury. Insight into the transcriptomic landscape of early cardiac regeneration from model organisms will shed light on evolutionarily conserved pathways in successful cardiac regeneration. METHODS: Here we describe a cross-species transcriptomic screen in 3 model organisms for cardiac regeneration: axolotl, neonatal mice, and zebrafish. Apical resection to remove ≈10% to 20% of ventricular mass was carried out in these model organisms. RNA-sequencing analysis was performed on the hearts harvested at 3 time points: 12, 24, and 48 hours after resection. Sham surgery was used as internal control. RESULTS: Genes associated with inflammatory processes were found to be upregulated in a conserved manner. Complement receptors (activated by complement components, part of the innate immune system) were found to be highly upregulated in all 3 species. This approach revealed induction of gene expression for complement 5a receptor 1 in the regenerating hearts of zebrafish, axolotls, and mice. Inhibition of complement 5a receptor 1 significantly attenuated the cardiomyocyte proliferative response to heart injury in all 3 species. Furthermore, after left ventricular apical resection, the cardiomyocyte proliferative response was diminished in mice with genetic deletion of complement 5a receptor 1. CONCLUSIONS: These data reveal that activation of complement 5a receptor 1 mediates an evolutionarily conserved response that promotes cardiomyocyte proliferation after cardiac injury and identify complement pathway activation as a common pathway of successful heart regeneration.


Asunto(s)
Evolución Molecular , Corazón/fisiología , Receptor de Anafilatoxina C5a/metabolismo , Regeneración/fisiología , Ambystoma mexicanum , Animales , Animales Recién Nacidos , Proliferación Celular , Perfilación de la Expresión Génica , Ontología de Genes , Ratones , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Péptidos Cíclicos/farmacología , ARN/química , ARN/aislamiento & purificación , ARN/metabolismo , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Análisis de Secuencia de ARN , Troponina T/análisis , Pez Cebra
3.
Respir Res ; 20(1): 133, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31262295

RESUMEN

BACKGROUND: Cystic fibrosis (CF) is an inherited disorder caused by mutations in the CF transmembrane conductance regulator (CFTR) gene that promotes persistent lung infection and inflammation and progressive loss of lung function. Patients with CF have increased lung lymphoid follicles (LFs) and B cell-activating factor of tumor necrosis factor family (BAFF) that regulates B cell survival and maturation. A direct role for CFTR in B cell activation and disease pathogenesis in CF remains unclear. METHODS: The number of LFs, BAFF+, TLR4+ and proliferation marker Ki67+ B cells in lung explants or resections from subjects with CF and normal controls was quantified by immunostaining. The role of CFTR in B cell activation and LF development was then examined in two independent cohorts of uninfected CFTR-deficient mice (Cftr -/-) and wild type controls. The number of lung LFs, B cells and BAFF+, CXCR4+, immunoglobulin G+ B cells was examined by immunostaining. Lung and splenocyte B cell activation marker and major histocompatibility complex class II (MHC class II) expression was quantified by flow cytometry. Inflammatory cytokine levels were measured in supernatants from isolated B cells from Cftr -/- and wild type mice stimulated in vitro with Pseudomonas aeruginosa lipopolysaccharide (LPS). RESULTS: There was a significant increase in well-formed LFs in subjects with CF compared to normal controls. Increased B cell activation and proliferation was observed in lung LFs from CF subjects as was quantified by a significant increase in B cell BAFF, TLR4 and Ki67 expression. Uninfected Cftr -/- mice had increased lung LFs and BAFF+ and CXCR4+ B cells compared to wild type controls. Lung B cells isolated from uninfected Cftr -/- mice demonstrated increased MHC class II expression. In vitro, isolated B cells from Cftr -/- mice produced increased IL-6 when stimulated with LPS compared to wild type controls. CONCLUSIONS: These data support a direct role for CFTR in B cell activation, proliferation and inflammatory cytokine production that promotes lung LF follicle development in cystic fibrosis.


Asunto(s)
Linfocitos B/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Fibrosis Quística/metabolismo , Estructuras Linfoides Terciarias/metabolismo , Adolescente , Animales , Fibrosis Quística/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Immunol ; 197(10): 4021-4033, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798158

RESUMEN

Substance P neuropeptide and its receptor, neurokinin-1 receptor (NK1R), are reported to present on the ocular surface. In this study, mice lacking functional NK1R exhibited an excessive desquamation of apical corneal epithelial cells in association with an increased epithelial cell proliferation and increased epithelial cell density, but decreased epithelial cell size. The lack of NK1R also resulted in decreased density of corneal nerves, corneal epithelial dendritic cells (DCs), and a reduced volume of basal tears. Interestingly, massive accumulation of CD11c+CD11b+ conventional DCs was noted in the bulbar conjunctiva and near the limbal area of corneas from NK1R-/- mice. After ocular HSV-1 infection, the number of conventional DCs and neutrophils infiltrating the infected corneas was significantly higher in NK1R-/- than C57BL/6J mice. This was associated with an increased viral load in infected corneas of NK1R-/- mice. As a result, the number of IFN-γ-secreting virus-specific CD4 T cells in the draining lymph nodes of NK1R-/- mice was much higher than in infected C57BL/6J mice. An increased number of CD4 T cells and mature neutrophils (CD11b+Ly6ghigh) in the inflamed corneas of NK1R-/- mice was associated with an early development of severe herpes stromal keratitis. Collectively, our results show that the altered corneal biology of uninfected NK1R-/- mice along with an enhanced immunological response after ocular HSV-1 infection causes an early development of herpes stromal keratitis in NK1R-/- mice.


Asunto(s)
Córnea/inmunología , Córnea/patología , Herpesvirus Humano 1/inmunología , Queratitis Herpética/virología , Receptores de Neuroquinina-1/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Conjuntiva/inmunología , Conjuntiva/patología , Conjuntiva/virología , Córnea/virología , Células Dendríticas/inmunología , Herpesvirus Humano 1/fisiología , Homeostasis , Interferón gamma/inmunología , Queratitis Herpética/inmunología , Queratitis Herpética/fisiopatología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptores de Neuroquinina-1/deficiencia , Receptores de Neuroquinina-1/inmunología , Carga Viral
5.
Am J Respir Cell Mol Biol ; 57(6): 711-720, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28708422

RESUMEN

Cystic fibrosis (CF) remains the most lethal genetic disease in the Caucasian population. However, there is great variability in clinical phenotypes and survival times, even among patients harboring the same genotype. We identified five patients with CF and a homozygous F508del mutation in the CFTR gene who were in their fifth or sixth decade of life and had shown minimal changes in lung function over a longitudinal period of more than 20 years. Because of the rarity of this long-term nonprogressive phenotype, we hypothesized these individuals may carry rare genetic variants in modifier genes that ameliorate disease severity. Individuals at the extremes of survival time and lung-function trajectory underwent whole-exome sequencing, and the sequencing data were filtered to include rare missense, stopgain, indel, and splicing variants present with a mean allele frequency of <0.2% in general population databases. Epithelial sodium channel (ENaC) mutants were generated via site-directed mutagenesis and expressed for Xenopus oocyte assays. Four of the five individuals carried extremely rare or never reported variants in the SCNN1D and SCNN1B genes of the ENaC. Separately, an independently enriched rare variant in SCNN1D was identified in the Exome Variant Server database associated with a milder pulmonary disease phenotype. Functional analysis using Xenopus oocytes revealed that two of the three variants in δ-ENaC encoded by SCNN1D exhibited hypomorphic channel activity. Our data suggest a potential role for δ-ENaC in controlling sodium reabsorption in the airways, and advance the plausibility of ENaC as a therapeutic target in CF.


Asunto(s)
Secuencia de Aminoácidos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Canales Epiteliales de Sodio/metabolismo , Eliminación de Secuencia , Animales , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Canales Epiteliales de Sodio/genética , Femenino , Humanos , Masculino , Xenopus , Xenopus laevis
6.
J Immunol ; 195(3): 1034-43, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26091719

RESUMEN

Staphylococcus aureus is well adapted to the human host. Evasion of the host phagocyte response is critical for successful infection. The staphylococcal bicomponent pore-forming toxins Panton-Valentine leukocidin LukSF-PV (PVL) and γ-hemolysin CB (HlgCB) target human phagocytes through interaction with the complement receptors C5aR1 and C5aR2. Currently, the apparent redundancy of both toxins cannot be adequately addressed in experimental models of infection because mice are resistant to PVL and HlgCB. The molecular basis for species specificity of the two toxins in animal models is not completely understood. We show that PVL and HlgCB feature distinct activity toward neutrophils of different mammalian species, where activity of PVL is found to be restricted to fewer species than that of HlgCB. Overexpression of various mammalian C5a receptors in HEK cells confirms that cytotoxicity toward neutrophils is driven by species-specific interactions of the toxins with C5aR1. By taking advantage of the species-specific engagement of the toxins with their receptors, we demonstrate that PVL and HlgCB differentially interact with human C5aR1 and C5aR2. In addition, binding studies illustrate that different parts of the receptor are involved in the initial binding of the toxin and the subsequent formation of lytic pores. These findings allow a better understanding of the molecular mechanism of pore formation. Finally, we show that the toxicity of PVL, but not of HlgCB, is neutralized by various C5aR1 antagonists. This study offers directions for the development of improved preclinical models for infection, as well as for the design of drugs antagonizing leukocidin toxicity.


Asunto(s)
Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Exotoxinas/inmunología , Proteínas Hemolisinas/inmunología , Leucocidinas/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Quimiocina/inmunología , Secuencia de Aminoácidos , Animales , Bovinos , Línea Celular , Células HEK293 , Humanos , Evasión Inmune/inmunología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neutrófilos/inmunología , Fagocitos/inmunología , Unión Proteica , Estructura Terciaria de Proteína , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptores de Quimiocina/antagonistas & inhibidores , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/patogenicidad
7.
Am J Respir Cell Mol Biol ; 55(5): 657-666, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27285858

RESUMEN

LPS-induced lung injury in the mouse is one of the most robust experimental models used for studies of acute lung injury (ALI) and acute respiratory distress syndrome in humans. Prior clinical and experimental studies support an important role for complement activation, particularly production of C5a, in the pathophysiology of human ALI/acute respiratory distress syndrome. In the mouse model, however, the precise role of C5a and its receptors is unclear. C5L2, an enigmatic second receptor for C5a, has been characterized, and results have generated substantial debate regarding its in vivo function. Our previous work with human neutrophils revealed a unique role for C5L2 in negatively modulating C5a-C5a receptor (C5aR)-mediated cellular activation, in which antibody-mediated blockade of C5L2 resulted in augmented C5a-C5aR responses. Here, we demonstrate that C5L2-/- mice (BALB/c background) administered intranasal LPS exhibit significantly more airway edema and hemorrhage than do wild-type animals. Bronchoalveolar lavage fluid and lung homogenates have significantly more neutrophils and myeloperoxidase activity, as well as proinflammatory cytokines and chemokines. When a blocking antibody against the C5aR was administered before LPS administration, the increased neutrophilic infiltration and cytokine levels were reversed. Thus, our data show not only that C5a contributes significantly to LPS-induced ALI in the mouse, but also that C5L2 plays an important antiinflammatory role in this model through actions resulting at least in part from negative modulation of C5a receptor activation.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Receptores de Quimiocina/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/fisiopatología , Administración Intranasal , Animales , Líquido del Lavado Bronquioalveolar , Quimiocinas/genética , Quimiocinas/metabolismo , Edema/complicaciones , Edema/patología , Edema/fisiopatología , Regulación de la Expresión Génica , Hemorragia/complicaciones , Hemorragia/patología , Hemorragia/fisiopatología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/administración & dosificación , Pulmón/patología , Ratones Endogámicos BALB C , Neutrófilos/metabolismo , Peroxidasa/metabolismo , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/deficiencia , Mecánica Respiratoria , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
8.
J Immunol ; 191(8): 4001-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043888

RESUMEN

The complement anaphylatoxin C5a is a critical mediator of allergic contact dermatitis, bridging essential aspects of innate and adaptive immunity. This anaphylatoxin functions by interacting with two 7-transmembrane segment receptors, the C5aR and C5L2. The C5aR is a classical G protein coupled receptor, whereas C5L2 is deficient in coupling to G proteins because of variations in the sequence. Our previous work in human neutrophils revealed a unique role for C5L2 in negatively modulating anaphylatoxin receptor mediated cellular activation through interactions with ß-arrestin. When C5L2 is deficient, C5aR-mediated ß-arrestin signaling is greatly enhanced. The work described in this study was undertaken first to determine the effect of C5L2 deficiency in a murine model of contact sensitivity, and second to determine whether the resultant exacerbation of inflammatory parameters reflects a negative modulatory function of C5L2 on the C5aR. First, we find dramatic increases in inflammation in C5L2(-/-) animals compared with wild type mice. Second, these increases are completely reversed following administration of mAb against the C5aR. Thus, in allergic contact sensitivity, as in isolated human neutrophils, C5L2 functions to suppress C5a-C5aR-mediated responses, further underscoring its role as a negative regulator of anaphylatoxin activity.


Asunto(s)
Complemento C5a/inmunología , Dermatitis Alérgica por Contacto/inmunología , Inflamación/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Quimiocina/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Arrestinas/inmunología , Arrestinas/metabolismo , Femenino , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Transducción de Señal , beta-Arrestinas
9.
J Immunol ; 190(9): 4836-47, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23547118

RESUMEN

According to the current paradigm, lymphocyte homing to the small intestine requires the expression of two tissue-specific homing receptors, the integrin α4ß7 and the CCL25 receptor CCR9. In this study, we investigated the organ distribution and the homing molecule expression of IgA Ab-secreting cells (ASCs) induced by intrarectal immunization with a particulate Ag, in comparison with other mucosal immunization routes. Intrarectal immunization induces gut-homing IgA ASCs that localize not only in the colon but also in the small intestine, although they are not responsive to CCL25, unlike IgA ASCs induced by oral immunization. The mucosal epithelial chemokine CCL28, known to attract all IgA ASCs, does not compensate for the lack of CCL25 responsiveness, because the number of Ag-specific cells is not decreased in the gut of CCR10-deficient mice immunized by the intrarectal route. However, Ag-specific IgA ASCs induced by intrarectal immunization express the integrin α4ß7, and their number is considerably decreased in the gut of ß7-deficient mice immunized by the intrarectal route, indicating that α4ß7 enables these cells to migrate into the small intestine, even without CCL25 responsiveness. In contrast, IgA ASCs induced by intranasal immunization express low α4ß7 levels and are usually excluded from the gut. Paradoxically, after intranasal immunization, Ag-specific IgA ASCs are significantly increased in the small intestine of ß7-deficient mice, demonstrating that lymphocyte homing is a competitive process and that integrin α4ß7 determines not only the intestinal tropism of IgA ASCs elicited in GALTs but also the intestinal exclusion of lymphocytes primed in other inductive sites.


Asunto(s)
Células Productoras de Anticuerpos/inmunología , Inmunoglobulina A/inmunología , Intestino Delgado/inmunología , Administración Rectal , Animales , Células Productoras de Anticuerpos/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Quimiocinas CC/inmunología , Quimiocinas CC/metabolismo , Colon/inmunología , Colon/metabolismo , Femenino , Inmunización/métodos , Inmunoglobulina A/metabolismo , Integrinas/inmunología , Integrinas/metabolismo , Intestino Delgado/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mucoproteínas , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Receptores CCR10/inmunología , Receptores CCR10/metabolismo
10.
J Am Soc Nephrol ; 25(2): 225-31, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24179165

RESUMEN

Necrotizing and crescentic GN (NCGN) with a paucity of glomerular immunoglobulin deposits is associated with ANCA. The most common ANCA target antigens are myeloperoxidase (MPO) and proteinase 3. In a manner that requires activation of the alternative complement pathway, passive transfer of antibodies to mouse MPO (anti-MPO) induces a mouse model of ANCA NCGN that closely mimics human disease. Here, we confirm the importance of C5aR/CD88 in the mediation of anti-MPO-induced NCGN and report that C6 is not required. We further demonstrate that deficiency of C5a-like receptor (C5L2) has the reverse effect of C5aR/CD88 deficiency and results in more severe disease, indicating that C5aR/CD88 engagement enhances inflammation and C5L2 engagement suppresses inflammation. Oral administration of CCX168, a small molecule antagonist of human C5aR/CD88, ameliorated anti-MPO-induced NCGN in mice expressing human C5aR/CD88. These observations suggest that blockade of C5aR/CD88 might have therapeutic benefit in patients with ANCA-associated vasculitis and GN.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/prevención & control , Autoantígenos/inmunología , Glomerulonefritis/prevención & control , Peroxidasa/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Administración Oral , Animales , Complemento C6/inmunología , Vía Alternativa del Complemento , Relación Dosis-Respuesta a Droga , Técnicas de Sustitución del Gen , Glomerulonefritis/complicaciones , Glomerulonefritis/inmunología , Hematuria/etiología , Hematuria/prevención & control , Humanos , Inmunización Pasiva , Leucocitos , Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/deficiencia , Proteinuria/etiología , Proteinuria/prevención & control , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores de Quimiocina/fisiología , Proteínas Recombinantes de Fusión , Orina/citología
11.
J Biol Chem ; 288(49): 35039-48, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24145037

RESUMEN

Chemokines display considerable promiscuity with multiple ligands and receptors shared in common, a phenomenon that is thought to underlie their biochemical "redundancy." Their receptors are part of a larger seven-transmembrane receptor superfamily, commonly referred to as G protein-coupled receptors, which have been demonstrated to be able to signal with different efficacies to their multiple downstream signaling pathways, a phenomenon referred to as biased agonism. Biased agonism has been primarily reported as a phenomenon of synthetic ligands, and the biologic prevalence and importance of such signaling are unclear. Here, to assess the presence of biased agonism that may underlie differential signaling by chemokines targeting the same receptor, we performed a detailed pharmacologic analysis of a set of chemokine receptors with multiple endogenous ligands using assays for G protein signaling, ß-arrestin recruitment, and receptor internalization. We found that chemokines targeting the same receptor can display marked differences in their efficacies for G protein- or ß-arrestin-mediated signaling or receptor internalization. This ligand bias correlates with changes in leukocyte migration, consistent with different mechanisms underlying the signaling downstream of these receptors induced by their ligands. These findings demonstrate that biased agonism is a common and likely evolutionarily conserved biological mechanism for generating qualitatively distinct patterns of signaling via the same receptor in response to different endogenous ligands.


Asunto(s)
Receptores de Quimiocina/agonistas , Receptores de Quimiocina/metabolismo , Arrestinas/metabolismo , Quimiocinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Cinética , Ligandos , Modelos Biológicos , Transducción de Señal , beta-Arrestinas
12.
J Allergy Clin Immunol ; 131(2): 541-8.e1-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22728083

RESUMEN

BACKGROUND: Mast cells express receptors for complement anaphylatoxins C3a and C5a (ie, C3a receptor [C3aR] and C5a receptor [C5aR]), and C3a and C5a are generated during various IgE-dependent immediate hypersensitivity reactions in vivo. However, it is not clear to what extent mast cell expression of C3aR or C5aR influences C3a- or C5a-induced cutaneous responses or IgE-dependent mast cell activation and passive cutaneous anaphylaxis (PCA) in vivo. OBJECTIVE: We sought to assess whether mouse skin mast cell expression of C3aR or C5aR influences (1) the cells' responsiveness to intradermal injections of C3a or C5a or (2) the extent of IgE-dependent mast cell degranulation and PCA in vivo. METHODS: We measured the magnitude of cutaneous responses to intradermal injections of C3a or C5a and the extent of IgE-dependent mast cell degranulation and PCA responses in mice containing mast cells that did or did not express C3aR or C5aR. RESULTS: The majority of the skin swelling induced by means of intradermal injection of C3a or C5a required that mast cells at the site expressed C3aR or C5aR, respectively, and the extent of IgE-dependent degranulation of skin mast cells and IgE-dependent PCA was significantly reduced when mast cells lacked either C3aR or C5aR. IgE-dependent PCA responses associated with local increases in C3a levels occurred in antibody-deficient mice but not in mice deficient in FcɛRIγ. CONCLUSION: Expression of C3aR and C5aR by skin mast cells contributes importantly to the ability of C3a and C5a to induce skin swelling and can enhance mast cell degranulation and inflammation during IgE-dependent PCA in vivo.


Asunto(s)
Inmunoglobulina E/metabolismo , Inflamación/inmunología , Mastocitos/inmunología , Receptor de Anafilatoxina C5a/biosíntesis , Receptores de Complemento/biosíntesis , Piel/inmunología , Anafilatoxinas/genética , Anafilatoxinas/inmunología , Animales , Células Cultivadas , Complemento C3a/genética , Complemento C3a/inmunología , Complemento C3a/metabolismo , Complemento C5a/genética , Complemento C5a/inmunología , Complemento C5a/metabolismo , Femenino , Inmunoglobulina E/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Piel/metabolismo , Piel/patología
13.
J Immunol ; 187(8): 4245-55, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21918196

RESUMEN

Respiratory syncytial virus (RSV) infection is associated with serious lung disease in infants and immunocompromised individuals and is linked to development of asthma. In mice, acute RSV infection causes airway hyperresponsiveness (AHR), inflammation, and mucus hypersecretion. Infected cells induce complement activation, producing the anaphylatoxin C3a. In this paper, we show RSV-infected wild-type mice produce Th17 cytokines, a response not previously associated with viral infections. Mice deficient in the C3aR fail to develop AHR following acute RSV infection, and production of Th17 cytokines was significantly attenuated. Tachykinin production also has been implicated in RSV pathophysiology, and tachykinin receptor-null mice were similarly protected from developing AHR. These animals were also deficient in production of Th17 cytokines. Tachykinin release was absent in mice deficient in C3aR, whereas C3a levels were unchanged in tachykinin receptor-null animals. Thus, our data reveal a crucial sequence following acute RSV infection where initial C3a production causes tachykinin release, followed by activation of the IL-17A pathway. Deficiency of either receptor affords protection from AHR, identifying two potential therapeutic targets.


Asunto(s)
Hiperreactividad Bronquial/inmunología , Complemento C3a/inmunología , Interleucina-17/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Taquicininas/inmunología , Animales , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/virología , Separación Celular , Complemento C3a/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Interleucina-17/metabolismo , Ratones , Ratones Noqueados , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Taquicininas/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(2): 628-32, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20018651

RESUMEN

Ubiquitously expressed seven-transmembrane receptors (7TMRs) classically signal through heterotrimeric G proteins and are commonly referred to as G protein-coupled receptors. It is now recognized that 7TMRs also signal through beta-arrestins, which act as versatile adapters controlling receptor signaling, desensitization, and trafficking. Most endogenous receptors appear to signal in a balanced fashion using both beta-arrestin and G protein-mediated pathways. Some 7TMRs are thought to be nonsignaling "decoys" because of their inability to activate typical G protein signaling pathways; it has been proposed that these receptors act to scavenge ligands or function as coreceptors. Here we demonstrate that ligand binding to the decoy receptor CXCR7 does not result in activation of signaling pathways typical of G proteins but does activate MAP kinases through beta-arrestins in transiently transfected cells. Furthermore, we observe that vascular smooth muscle cells that endogenously express CXCR7 migrate to its ligand interferon-inducible T-cell alpha chemoattractant (ITAC), an effect that is significantly attenuated by treatment with either a CXCR7 antagonist or beta-arrestin depletion by siRNA. This example of an endogenous "beta-arrestin-biased" 7TMR that signals through beta-arrestin in the absence of G protein activation demonstrates that some 7TMRs encoded in the genome have evolved to signal through beta-arrestin exclusively and suggests that other receptors that are currently thought to be orphans or decoys may also signal through such nonclassical pathways.


Asunto(s)
Arrestinas/fisiología , Receptores CXCR/fisiología , Linfocitos T/fisiología , Animales , Línea Celular , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/fisiología , Humanos , Ratones , Músculo Liso Vascular/fisiología , Fosforilación , ARN Mensajero/genética , Ratas , Receptores CXCR/genética , Receptores de Quimiocina/genética , Transducción de Señal/fisiología , Linfocitos T/citología , beta-Arrestinas
15.
J Immunol ; 185(11): 6741-52, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20974988

RESUMEN

The complement fragment C5a plays dual roles in the development of experimental allergic asthma. It protects from pulmonary allergy by a regulatory effect on dendritic cells during allergen sensitization, but is proallergic during the effector phase. C5a can bind to two distinct receptors (i.e., C5a receptor and C5a receptor-like 2 [C5L2]). The functional role of C5L2 in vivo remains enigmatic. In this study, we show in two models of OVA- and house dust mite (HDM)-induced experimental allergic asthma that C5L2-deficient mice are protected from the development of airway hyperresponsiveness, Th2 cytokine production, eosinophilic airway inflammation, serum IgE, or mucus production. Surprisingly, HDM-induced experimental asthma in C5L2-deficient mice was associated with increased pulmonary IL-17A production and increased airway neutrophil numbers. To directly assess the role of C5L2 on myeloid dendritic cells (mDCs) during allergen sensitization, we performed single or repeated adoptive transfers of C5L2-deficient mDCs into wild-type mice. HDM-pulsed C5L2-deficient mDCs induced strong Th2 cytokine production, which was associated with marked IFN-γ and IL-17A production, decreased eosinophil numbers, and reduced IgE production as compared with HDM-pulsed mDCs from wild-type mice. HDM stimulation of C5L2(-/-) mDCs in vitro resulted in production of Th17-promoting cytokine IL-23, which was absent in wild-type mDCs. Our findings suggest that C5L2 acts at the mDC/T cell interface to control the development of Th1 and Th17 cells in response to airway HDM exposure. Furthermore, it drives Th2 immune responses independent of mDCs, suggesting a complex role for C5L2 in the development of experimental allergic asthma.


Asunto(s)
Alérgenos/inmunología , Asma/inmunología , Asma/metabolismo , Receptores de Quimiocina/fisiología , Alérgenos/administración & dosificación , Alérgenos/toxicidad , Animales , Asma/patología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/prevención & control , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Polvo/inmunología , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/toxicidad , Pyroglyphidae/inmunología , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/patología
16.
J Biol Chem ; 285(10): 7633-44, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20044484

RESUMEN

The complement anaphylatoxin C5a is a proinflammatory component of host defense that functions through two identified receptors, C5a receptor (C5aR) and C5L2. C5aR is a classical G protein-coupled receptor, whereas C5L2 is structurally homologous but deficient in G protein coupling. In human neutrophils, we show C5L2 is predominantly intracellular, whereas C5aR is expressed on the plasma membrane. Confocal analysis shows internalized C5aR following ligand binding is co-localized with both C5L2 and beta-arrestin. Antibody blockade of C5L2 results in a dramatic increase in C5a-mediated chemotaxis and ERK1/2 phosphorylation but does not alter C5a-mediated calcium mobilization, supporting its role in modulation of the beta-arrestin pathway. Association of C5L2 with beta-arrestin is confirmed by cellular co-immunoprecipitation assays. C5L2 blockade also has no effect on ligand uptake or C5aR endocytosis in human polymorphonuclear leukocytes, distinguishing its role from that of a rapid recycling or scavenging receptor in this cell type. This is thus the first example of a naturally occurring seven-transmembrane segment receptor that is both obligately uncoupled from G proteins and a negative modulator of signal transduction through the beta-arrestin pathway. Physiologically, these properties provide the possibility for additional fine-tuning of host defense.


Asunto(s)
Receptor de Anafilatoxina C5a/metabolismo , Receptores de Quimiocina/metabolismo , Transducción de Señal/fisiología , Animales , Arrestinas/metabolismo , Línea Celular , Quimiotaxis de Leucocito/fisiología , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Neutrófilos/citología , Neutrófilos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor de Anafilatoxina C5a/química , Receptor de Anafilatoxina C5a/genética , Receptores de Quimiocina/química , Receptores de Quimiocina/genética , Distribución Tisular , beta-Arrestinas
17.
J Cell Biochem ; 112(10): 2759-72, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21748788

RESUMEN

Heterotopic ossification (HO) is a disabling condition associated with neurologic injury, inflammation, and overactive bone morphogenetic protein (BMP) signaling. The inductive factors involved in lesion formation are unknown. We found that the expression of the neuro-inflammatory factor Substance P (SP) is dramatically increased in early lesional tissue in patients who have either fibrodysplasia ossificans progressiva (FOP) or acquired HO, and in three independent mouse models of HO. In Nse-BMP4, a mouse model of HO, robust HO forms in response to tissue injury; however, null mutations of the preprotachykinin (PPT) gene encoding SP prevent HO. Importantly, ablation of SP(+) sensory neurons, treatment with an antagonist of SP receptor NK1r, deletion of NK1r gene, or genetic down-regulation of NK1r-expressing mast cells also profoundly inhibit injury-induced HO. These observations establish a potent neuro-inflammatory induction and amplification circuit for BMP-dependent HO lesion formation, and identify novel molecular targets for prevention of HO.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Osificación Heterotópica/metabolismo , Sustancia P/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Morfogenéticas Óseas/genética , Femenino , Humanos , Inmunohistoquímica , Isoindoles/farmacología , Masculino , Ratones , Ratones Transgénicos , Miositis Osificante/genética , Miositis Osificante/metabolismo , Antagonistas del Receptor de Neuroquinina-1 , Osificación Heterotópica/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores de Neuroquinina-1/metabolismo , Células Receptoras Sensoriales/metabolismo , Taquicininas/genética , Taquicininas/metabolismo
18.
Am J Pathol ; 174(3): 782-96, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19234135

RESUMEN

Neprilysin is a transmembrane metalloendopeptidase that degrades neuropeptides that are important for both growth and contraction. In addition to promoting carcinogenesis, decreased levels of neprilysin increases inflammation and neuroendocrine cell hyperplasia, which may predispose to vascular remodeling. Early pharmacological studies showed a decrease in chronic hypoxic pulmonary hypertension with neprilysin inhibition. We used a genetic approach to test the alternate hypothesis that neprilysin depletion increases chronic hypoxic pulmonary hypertension. Loss of neprilysin had no effect on baseline airway or alveolar wall architecture, vessel density, cardiac function, hematocrit, or other relevant peptidases. Only lung neuroendocrine cell hyperplasia and a subtle neuropeptide imbalance were found. After chronic hypoxia, neprilysin-null mice exhibited exaggerated pulmonary hypertension and striking increases in muscularization of distal vessels. Subtle thickening of proximal media/adventitia not typically seen in mice was also detected. In contrast, adaptive right ventricular hypertrophy was less than anticipated. Hypoxic wild-type pulmonary vessels displayed close temporal and spatial relationships between decreased neprilysin and increased cell growth. Smooth muscle cells from neprilysin-null pulmonary arteries had increased proliferation compared with controls, which was decreased by neprilysin replacement. These data suggest that neprilysin may be protective against chronic hypoxic pulmonary hypertension in the lung, at least in part by attenuating the growth of smooth muscle cells. Lung-targeted strategies to increase neprilysin levels could have therapeutic benefits in the treatment of this disorder.


Asunto(s)
Hipertensión Pulmonar/patología , Hipoxia/genética , Ratones Noqueados , Neprilisina/deficiencia , Arteria Pulmonar/patología , Circulación Pulmonar/fisiología , Animales , División Celular , Enfermedad Crónica , Cartilla de ADN , Predisposición Genética a la Enfermedad , Genotipo , Hemodinámica , Hipertensión Pulmonar/genética , Hipoxia/patología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/patología , Neprilisina/genética
19.
Immunobiology ; 225(2): 151895, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31843260

RESUMEN

Signal transduction by the Toll-like receptors (TLRs) is a key component of innate immunity against many pathogens and also underlies a large burden of human diseases. Therefore, the mechanisms and regulation of signaling from the TLRs are of considerable interest. Here we seek to determine the molecular mechanism by which TLR2 and TLR4, members of the Toll-like receptor family, are activated by bacterial LPS, hyperoxia, and zymosan respectively. Our central hypothesis is that the oxidation state of cysteine thiols on the ectodomain of TLR2 and TLR4 are critical for pathogen-initiated intracellular signaling as well in hyperoxia. Cysteine thiols of TLR4 and its co-receptor MD2 have been shown to aid binding between the two molecules and also bacterial LPS binding to the receptor complex. We extend these findings by demonstrating the oxidation of free thiols on the ectodomain of hTLR4, after exposure to LPS or hyperoxia suggesting that the cysteines on the ectodomain of TLR4 could form intra- or intermolecular disulfide bonds. We also demonstrated blockade of intracellular signaling from TLR4 and TLR2 by thiol-modifying compounds which suggest a novel therapeutic intervention for sepsis, hyperoxia-induced cell injury and yeast infection. In these experiments CHO-3E10, HEK293 cells expressing hTLR2 or hTLR4 and mouse peritoneal macrophages cells were pretreated with cell impermeable maleimides to alkylate thiols on the extracellular domain of TLRs, cells were then exposed to LPS, hyperoxia or zymosan. In all of these models, we detected decreased intracellular signaling from TLR2 or TLR4. Furthermore, incubation with phenyl arsine oxide - which forms stable complexes with vicinal cysteine residues - prevented LPS induced HEK293/hTLR4 intracellular signaling which was reversed by DMPS. Sequence analysis of different TLRs revealed Leucine-Rich Repeat C-terminal (LRRCT) domain that contains 4 conserved cysteines. Further work is required to pinpoint the role of each cysteine in receptor dimerization, pathogen binding, hyperoxia modulation, and intracellular signaling.


Asunto(s)
Cisteína/metabolismo , Transducción de Señal/fisiología , Compuestos de Sulfhidrilo/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células CHO , Línea Celular , Cricetulus , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C3H , Oxidación-Reducción/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
20.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L687-97, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19633070

RESUMEN

The neuropeptide substance P manifests its biological functions through ligation of a G protein-coupled receptor, the NK1R. Mice with targeted deletion of this receptor reveal a preponderance of proinflammatory properties resulting from ligand activation, demonstrating a neurogenic component to multiple forms of inflammation and injury. We hypothesized that NK1R deficiency would afford a similar protection from inflammation associated with hyperoxia. Counter to our expectations, however, NK1R-/- animals suffered significantly worse lung injury compared with wild-type mice following exposure to 90% oxygen. Median survival was shortened to 84 h for NK1R-/- mice from 120 h for wild-type animals. Infiltration of inflammatory cells into the lungs was significantly increased; NK1R-/- animals also exhibited increased pulmonary edema, hemorrhage, and bronchoalveolar lavage fluid protein levels. TdT-mediated dUTP nick end labeling (TUNEL) staining was significantly elevated in NK1R-/- animals following hyperoxia. Furthermore, induction of metallothionein and Na(+)-K(+)-ATPase was accelerated in NK1R-/- compared with wild-type mice, consistent with increased oxidative injury and edema. In cultured mouse lung epithelial cells in 95% O(2), however, addition of substance P promoted cell death, suggesting the neurogenic component of hyperoxic lung injury is mediated by additional mechanisms in vivo. Release of bioactive constituents including substance P from sensory neurons results from activation of the vanilloid receptor, TRPV1. In mice with targeted deletion of the TRPV1 gene, acute hyperoxic injury is attenuated relative to NK1R-/- animals. Our findings thus reveal a major neurogenic mechanism in acute hyperoxic lung injury and demonstrate concerted actions of sensory neurotransmitters revealing significant protection for NK1R-mediated functions.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/prevención & control , Hiperoxia , Oxígeno/metabolismo , Receptores de Neuroquinina-1/fisiología , Lesión Pulmonar Aguda/patología , Animales , Apoptosis , Western Blotting , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Edema/etiología , Edema/metabolismo , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Peroxidasa/metabolismo , Tasa de Supervivencia , Canales Catiónicos TRPV/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA