Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 523(7560): 342-6, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26053123

RESUMEN

Lipid mediators influence immunity in myriad ways. For example, circulating sphingosine-1-phosphate (S1P) is a key regulator of lymphocyte egress. Although the majority of plasma S1P is bound to apolipoprotein M (ApoM) in the high-density lipoprotein (HDL) particle, the immunological functions of the ApoM-S1P complex are unknown. Here we show that ApoM-S1P is dispensable for lymphocyte trafficking yet restrains lymphopoiesis by activating the S1P1 receptor on bone marrow lymphocyte progenitors. Mice that lacked ApoM (Apom(-/-)) had increased proliferation of Lin(-) Sca-1(+) cKit(+) haematopoietic progenitor cells (LSKs) and common lymphoid progenitors (CLPs) in bone marrow. Pharmacological activation or genetic overexpression of S1P1 suppressed LSK and CLP cell proliferation in vivo. ApoM was stably associated with bone marrow CLPs, which showed active S1P1 signalling in vivo. Moreover, ApoM-bound S1P, but not albumin-bound S1P, inhibited lymphopoiesis in vitro. Upon immune stimulation, Apom(-/-) mice developed more severe experimental autoimmune encephalomyelitis, characterized by increased lymphocytes in the central nervous system and breakdown of the blood-brain barrier. Thus, the ApoM-S1P-S1P1 signalling axis restrains the lymphocyte compartment and, subsequently, adaptive immune responses. Unique biological functions imparted by specific S1P chaperones could be exploited for novel therapeutic opportunities.


Asunto(s)
Apolipoproteínas/metabolismo , Sistema Nervioso Central/patología , Lipoproteínas HDL/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Linfopoyesis , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Apolipoproteínas/deficiencia , Apolipoproteínas/genética , Apolipoproteínas M , Barrera Hematoencefálica/patología , Movimiento Celular , Proliferación Celular/genética , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Clorhidrato de Fingolimod/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Linfocitos/inmunología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Lisofosfolípidos/agonistas , Lisofosfolípidos/sangre , Lisofosfolípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/agonistas , Esfingosina/sangre , Esfingosina/genética , Esfingosina/metabolismo
2.
Proc Natl Acad Sci U S A ; 114(17): 4531-4536, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396408

RESUMEN

The vasculature of the central nervous system (CNS) forms a selective barrier termed the blood-brain barrier (BBB). Disruption of the BBB may contribute to various CNS diseases. Conversely, the intact BBB restricts efficient penetration of CNS-targeted drugs. Here, we report the BBB-regulatory role of endothelial sphingosine 1-phosphate (S1P) receptor-1, a G protein-coupled receptor known to promote the barrier function in peripheral vessels. Endothelial-specific S1pr1 knockout mice (S1pr1iECKO ) showed BBB breach for small-molecular-mass fluorescence tracers (<3 kDa), but not larger tracers (>10 kDa). Chronic BBB leakiness was associated with cognitive impairment, as assessed by the novel object recognition test, but not signs of brain inflammation. Brain microvessels of S1pr1iECKO mice showed altered subcellular distribution of tight junctional proteins. Pharmacological inhibition of S1P1 function led to transient BBB breach. These data suggest that brain endothelial S1P1 maintain the BBB by regulating the proper localization of tight junction proteins and raise the possibility that endothelial S1P1 inhibition may be a strategy for transient BBB opening and delivery of small molecules into the CNS.


Asunto(s)
Barrera Hematoencefálica/fisiología , Endotelio Vascular/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Transporte Biológico , Encéfalo/irrigación sanguínea , Células Endoteliales/fisiología , Regulación de la Expresión Génica , Lisofosfolípidos , Ratones , Ratones Noqueados , Receptores de Lisoesfingolípidos/genética , Esfingosina/análogos & derivados , Uniones Estrechas/metabolismo
3.
Gastroenterology ; 144(4): 771-80, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23313268

RESUMEN

BACKGROUND & AIMS: Glucose is absorbed into intestine cells via the sodium glucose transporter 1 (SGLT-1) and glucose transporter 2 (GLUT2); various peptides and hormones control this process. Apelin is a peptide that regulates glucose homeostasis and is produced by proximal digestive cells; we studied whether glucose modulates apelin secretion by enterocytes and the effects of apelin on intestinal glucose absorption. METHODS: We characterized glucose-related luminal apelin secretion in vivo and ex vivo by mass spectroscopy and immunologic techniques. The effects of apelin on (14)C-labeled glucose transport were determined in jejunal loops and in mice following apelin gavage. We determined levels of GLUT2 and SGLT-1 proteins and phosphorylation of AMPKα2 by immunoblotting. The net effect of apelin on intestinal glucose transepithelial transport was determined in mice. RESULTS: Glucose stimulated luminal secretion of the pyroglutaminated apelin-13 isoform ([Pyr-1]-apelin-13) in the small intestine of mice. Apelin increased specific glucose flux through the gastric epithelial barrier in jejunal loops and in vivo following oral glucose administration. Conversely, pharmacologic apelin blockade in the intestine reduced the increased glycemia that occurs following oral glucose administration. Apelin activity was associated with phosphorylation of AMPKα2 and a rapid increase of the GLUT2/SGLT-1 protein ratio in the brush border membrane. CONCLUSIONS: Glucose amplifies its own transport from the intestinal lumen to the bloodstream by increasing luminal apelin secretion. In the lumen, active apelin regulates carbohydrate flux through enterocytes by promoting AMPKα2 phosphorylation and modifying the ratio of SGLT-1:GLUT2. The glucose-apelin cycle might be pharmacologically handled to regulate glucose absorption and assess better control of glucose homeostasis.


Asunto(s)
Carbohidratos/farmacocinética , Glucosa/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Análisis de Varianza , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Western Blotting , Cromatografía Liquida/métodos , Modelos Animales de Enfermedad , Glucosa/farmacología , Transportador de Glucosa de Tipo 2/metabolismo , Inmunohistoquímica , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Distribución Aleatoria , Valores de Referencia , Transportador 1 de Sodio-Glucosa/metabolismo
4.
Proc Natl Acad Sci U S A ; 108(23): 9613-8, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21606363

RESUMEN

Protection of the endothelium is provided by circulating sphingosine-1-phosphate (S1P), which maintains vascular integrity. We show that HDL-associated S1P is bound specifically to both human and murine apolipoprotein M (apoM). Thus, isolated human ApoM(+) HDL contained S1P, whereas ApoM(-) HDL did not. Moreover, HDL in Apom(-/-) mice contains no S1P, whereas HDL in transgenic mice overexpressing human apoM has an increased S1P content. The 1.7-Å structure of the S1P-human apoM complex reveals that S1P interacts specifically with an amphiphilic pocket in the lipocalin fold of apoM. Human ApoM(+) HDL induced S1P(1) receptor internalization, downstream MAPK and Akt activation, endothelial cell migration, and formation of endothelial adherens junctions, whereas apoM(-) HDL did not. Importantly, lack of S1P in the HDL fraction of Apom(-/-) mice decreased basal endothelial barrier function in lung tissue. Our results demonstrate that apoM, by delivering S1P to the S1P(1) receptor on endothelial cells, is a vasculoprotective constituent of HDL.


Asunto(s)
Apolipoproteínas/metabolismo , Endotelio Vascular/metabolismo , Lipoproteínas HDL/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Apolipoproteínas/química , Apolipoproteínas/genética , Apolipoproteínas M , Western Blotting , Células Cultivadas , Cristalografía por Rayos X , Endocitosis , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Activación Enzimática , Células HEK293 , Humanos , Lipocalinas/química , Lipocalinas/genética , Lipocalinas/metabolismo , Lipoproteínas HDL/química , Lisofosfolípidos/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/química , Esfingosina/metabolismo
5.
Sci Signal ; 17(824): eadg9256, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38377179

RESUMEN

High-density lipoprotein (HDL) nanoparticles promote endothelial cell (EC) function and suppress inflammation, but their utility in treating EC dysfunction has not been fully explored. Here, we describe a fusion protein named ApoA1-ApoM (A1M) consisting of apolipoprotein A1 (ApoA1), the principal structural protein of HDL that forms lipid nanoparticles, and ApoM, a chaperone for the bioactive lipid sphingosine 1-phosphate (S1P). A1M forms HDL-like particles, binds to S1P, and is signaling competent. Molecular dynamics simulations showed that the S1P-bound ApoM moiety in A1M efficiently activated EC surface receptors. Treatment of human umbilical vein ECs with A1M-S1P stimulated barrier function either alone or cooperatively with other barrier-enhancing molecules, including the stable prostacyclin analog iloprost, and suppressed cytokine-induced inflammation. A1M-S1P injection into mice during sterile inflammation suppressed neutrophil influx and inflammatory mediator secretion. Moreover, systemic A1M administration led to a sustained increase in circulating HDL-bound S1P and suppressed inflammation in a murine model of LPS-induced endotoxemia. We propose that A1M administration may enhance vascular endothelial barrier function, suppress cytokine storm, and promote resilience of the vascular endothelium.


Asunto(s)
Apolipoproteínas , Lipocalinas , Humanos , Ratones , Animales , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacología , Lipocalinas/metabolismo , Lipocalinas/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Apolipoproteínas M , Inflamación , Lipoproteínas HDL/farmacología , Lipoproteínas HDL/metabolismo , Lisofosfolípidos/farmacología , Lisofosfolípidos/metabolismo , Esfingosina
7.
Circulation ; 124(24): 2725-34, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22082680

RESUMEN

BACKGROUND: Outcomes for organ transplantation are constantly improving because of advances in organ preservation, surgical techniques, immune clinical monitoring, and immunosuppressive treatment preventing acute transplant rejection. However, chronic rejection including transplant vasculopathy still limits long-term patient survival. Transplant vasculopathy is characterized by progressive neointimal hyperplasia leading to arterial stenosis and ischemic failure of the allograft. This work sought to decipher the manner in which the humoral immune response, mimicked by W6/32 anti-HLA antibody, contributes to transplant vasculopathy. METHODS AND RESULTS: Studies were performed in vitro on cultured human smooth muscle cells, ex vivo on human arterial segments, and in vivo in a model consisting of human arterial segments grafted into severe combined immunodeficiency/beige mice injected weekly with anti-HLA antibodies. We report that anti-HLA antibodies are mitogenic for smooth muscle cells through a signaling mechanism implicating matrix metalloproteinases (MMPs) (membrane type 1 MMP and MMP2) and neutral sphingomyelinase-2. This mitogenic signaling and subsequent DNA synthesis are blocked in smooth muscle cells silenced for MMP2 or for neutral sphingomyelinase-2 by small interfering RNAs, in smooth muscle cells transfected with a vector coding for a dominant-negative form of membrane type 1 MMP, and after treatment by pharmacological inhibitors of MMPs (Ro28-2653) or neutral sphingomyelinase-2 (GW4869). In vivo, Ro28-2653 and GW4869 reduced the intimal thickening induced by anti-HLA antibodies in human mesenteric arteries grafted into severe combined immunodeficiency/beige mice. CONCLUSIONS: These data highlight a crucial role for MMP2 and neutral sphingomyelinase-2 in vasculopathy triggered by a humoral immune response and open new perspectives for preventing transplant vasculopathy with the use of MMP and neutral sphingomyelinase inhibitors, in addition to conventional immunosuppression.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Arterias/trasplante , Antígenos HLA/inmunología , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Enfermedades Vasculares/fisiopatología , Compuestos de Anilina/farmacología , Animales , Anticuerpos Antiidiotipos/efectos adversos , Arterias/patología , Arterias/fisiopatología , Compuestos de Bencilideno/farmacología , Células Cultivadas , Constricción Patológica/etiología , Constricción Patológica/fisiopatología , Modelos Animales de Enfermedad , Humanos , Hiperplasia/etiología , Hiperplasia/fisiopatología , Técnicas In Vitro , Metaloproteinasa 14 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones SCID , Modelos Animales , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Neointima/patología , Neointima/fisiopatología , Piperazinas/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/efectos de los fármacos , Enfermedades Vasculares/etiología , Injerto Vascular
8.
Am J Pathol ; 174(1): 44-53, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19095959

RESUMEN

Treatment of cancer using radiation can be significantly compromised by the development of severe acute and late damage to normal tissue. Treatments that either reduce the risk and severity of damage or that facilitate the healing of radiation injuries are being developed, including autologous adipose tissue grafts to repair tissue defects or involutional disorders that result from tumor resection. Adipose tissue is specialized in energy storage and contains different cell types, including preadipocytes, which could be used for autologous transplantation. It has long been considered a poorly proliferative connective tissue; however, the acute effects of ionizing radiation on adipose tissue have not been investigated. Therefore, the aim of this study was to characterize the alterations induced in adipose tissue by total body irradiation. A severe decrease in proliferating cells, as well as a significant increase in apoptotic cells, was observed in vivo in inguinal fat pads following irradiation. Additionally, irradiation altered the hematopoietic population. Decreases in the proliferation and differentiation capacities of non-hematopoietic progenitors were also observed following irradiation. Together, these data demonstrate that subcutaneous adipose tissue is very sensitive to irradiation, leading to a profound alteration of its developmental potential. This damage could also alter the reconstructive properties of adipose tissue and, therefore, calls into question its use in autologous fat transfer following radiotherapy.


Asunto(s)
Adipocitos/efectos de la radiación , Tejido Adiposo/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Citometría de Flujo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Estrés Oxidativo/efectos de la radiación , Fenotipo , Reacción en Cadena de la Polimerasa , Traumatismos Experimentales por Radiación/metabolismo , Células Madre/metabolismo , Células Madre/patología , Células Madre/efectos de la radiación
9.
Cytokine ; 45(3): 169-73, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19147373

RESUMEN

T-cell functions are currently used as biomarkers for the pharmacodynamic monitoring of immunosuppressive drugs or as disease biomarkers of inflammation/sepsis and organ rejection. In order to evaluate co-factors potentially influencing the expression of the immunological biomarkers, we explored T-cell proliferation, T-cell activation (CD25 and CD71 expressions) and intra-lymphocyte cytokine production (interleukin (IL)-2 and tumor necrosis factor (TNF)-alpha) in healthy volunteers, dialysis patients and stable kidney-transplant patients treated with standard immunosuppressive therapy, i.e. tacrolimus, mycophenolic acid with or without steroids. Age was positively correlated with TNF-alpha expression in all three patient populations, and with IL-2 expression in healthy volunteers and kidney-transplant patients. Further age was correlated with inhibition of lymphocyte proliferation in healthy volunteers and with the T-cell activation marker CD25 in kidney-transplant patients. In healthy volunteers lymphocyte proliferation was higher in woman as compared to men. Other biomarkers of T-cell function were independent of the gender. In the kidney-transplant patient group a significantly lower expression of all biomarkers of T-cell functions compared to healthy volunteers and dialysis patients. In dialysis patients we found significant increased IL-2 expression compared to healthy volunteers, while the other T-cell functions were not significantly different. Further time on dialysis had no effect on the level of biomarker expression. In conclusion we found decreased T-cell functions in kidney-transplant patients compared to healthy volunteers and dialysis patients, increased IL-2 expression in dialysis patients compared to healthy volunteers and in all three populations we found a correlation of age and intra-T-lymphocyte TNF-alpha expression.


Asunto(s)
Biomarcadores/metabolismo , Citocinas/metabolismo , Trasplante de Riñón/inmunología , Diálisis Renal , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Adulto , Envejecimiento , Antígenos CD/biosíntesis , Proliferación Celular , Femenino , Humanos , Interleucina-2/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Masculino , Persona de Mediana Edad , Receptores de Transferrina/biosíntesis
10.
J Exp Med ; 216(7): 1582-1598, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31147448

RESUMEN

Sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) activate G protein-coupled receptors (GPCRs) to regulate biological processes. Using a genome-wide CRISPR/dCas9-based GPCR signaling screen, LPAR1 was identified as an inducer of S1PR1/ß-arrestin coupling while suppressing Gαi signaling. S1pr1 and Lpar1-positive lymphatic endothelial cells (LECs) of lymph nodes exhibit constitutive S1PR1/ß-arrestin signaling, which was suppressed by LPAR1 antagonism. Pharmacological inhibition or genetic loss of function of Lpar1 reduced the frequency of punctate junctions at sinus-lining LECs. Ligand activation of transfected LPAR1 in endothelial cells remodeled junctions from continuous to punctate structures and increased transendothelial permeability. In addition, LPAR1 antagonism in mice increased lymph node retention of adoptively transferred lymphocytes. These data suggest that cross-talk between LPAR1 and S1PR1 promotes the porous junctional architecture of sinus-lining LECs, which enables efficient lymphocyte trafficking. Heterotypic inter-GPCR coupling may regulate complex cellular phenotypes in physiological milieu containing many GPCR ligands.


Asunto(s)
Células Endoteliales/metabolismo , Ganglios Linfáticos/metabolismo , Receptor Cross-Talk , Animales , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Técnica del Anticuerpo Fluorescente , Edición Génica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lisofosfolípidos/metabolismo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
11.
Free Radic Biol Med ; 45(10): 1457-67, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18801426

RESUMEN

Reactive carbonyl compounds (RCC) generated by polyunsaturated fatty acid oxidation alter progressively cellular and tissular proteins by forming adducts on free amino groups and thiol residues (carbonyl stress). Carbonyl scavengers may neutralize RCC, but their protective effect in atherosclerosis has not been extensively studied. We report the carbonyl scavenger and antiatherogenic properties of hydrazine derivatives, namely hydralazine, an antihypertensive drug, isoniazid, an antituberculosis agent, and two antidepressants, phenelzine and iproniazid. These drugs were poorly efficient in preventing the oxidation of LDL mediated by smooth muscle cells (SMCs), but inhibited the toxicity of UV-oxidized LDL (oxLDL) and of 4-hydroxynonenal (4-HNE). Hydrazine derivatives prevented the formation of foam cells resulting from LDL oxidation in human macrophagic U937 cells, and blocked the carbonyl stress in SMCs, by inhibiting the decrease in free amino group content, the increase in carbonylated proteins, and the formation of 4-HNE adducts on PDGFR. Experimental studies carried out on apoE-/- mice supplemented with drugs (30 mg/L in drinking water) showed a significant carbonyl stress inhibition correlated with a net reduction of atherosclerotic lesion development. In conclusion, these data indicate that hydrazine derivatives exhibit carbonyl scavenger and antiatherogenic properties, which opens novel therapeutical approaches for atherosclerosis and its cardiovascular complications.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Depuradores de Radicales Libres/farmacología , Hidrazinas/farmacología , Animales , Apolipoproteínas E/deficiencia , Células Cultivadas , Citoprotección/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Depuradores de Radicales Libres/química , Humanos , Hidrazinas/química , Lipoproteínas/farmacología , Masculino , Ratones , Ratones Noqueados , Estructura Molecular , Conejos , Células U937
12.
Transpl Immunol ; 19(2): 112-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18503886

RESUMEN

The incidence of acute rejection is significantly higher in hepatitis C virus (HCV) liver-transplant patients than in patients who have received a graft for other liver diseases, i.e., mainly alcoholic cirrhosis. The aim of this study was to assess T-cell function, i.e., intralymphocyte cytokine expression (IL-2 and TNF-alpha), T-cell activation [i.e., transferrin receptor (CD71) and interleukin (IL)-2 alpha-chain (CD25) expression], and T-cell proliferation using a flow-cytometry whole-blood assay in patients waiting for a liver transplantation (n=49). Our data suggest that, in mitogen-stimulated T-cells, (i) intra-lymphocyte cytokine expression is significantly higher in patients with liver disease than in healthy volunteers (n=25); (ii) the expression of T-cell activation markers is decreased in patients with liver cirrhosis compared to healthy volunteers, and (iii) the expression of T-cell activation markers and T-cell proliferation are increased in patients with HCV infection (n=15) compared to those without HCV infection (n=34), particularly compared to patients with alcoholic liver disease (n=19). Circulating CD19-positive cells count was also significantly higher in HCV-positive patients. In conclusion, in vitro, mitogen-stimulated T-cell seem to induce a higher immune response in the blood from patients waiting for a liver transplant for HCV-related liver disease than those without HCV infection, and particularly those with alcoholic liver disease.


Asunto(s)
Citocinas/sangre , Hepatitis C Crónica/inmunología , Trasplante de Hígado/inmunología , Activación de Linfocitos , Mitógenos/metabolismo , Subgrupos de Linfocitos T/inmunología , Adulto , Anciano , Biomarcadores/sangre , Proliferación Celular , Femenino , Hepacivirus/inmunología , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/virología , Humanos , Hepatopatías/inmunología , Masculino , Persona de Mediana Edad , Mitógenos/inmunología , Subgrupos de Linfocitos T/metabolismo
13.
Int Immunopharmacol ; 8(5): 769-73, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18387521

RESUMEN

BACKGROUND: The formulations of mycophenolic acid, i.e., mycophenolate mofetil (MMF) and enteric-coated mycophenolate sodium (EC-MPS), seem to have different pharmacokinetic profiles. The aim of this study was to compare the effects MMF and EC-MPS on T-cell proliferation, T-cell activation, T-cell function, and lymphocyte subsets. CLINICAL STUDY AND METHODS: Ten stable kidney-transplant patients on standard maintenance therapy of tacrolimus and MMF (1 g/d), with or without steroids, were converted from MMF to EC-MPS at equivalent dose (720 mg/d). Tacrolimus and steroid doses remained unchanged before, and at 1, 2, 3, and 6 months (M) after conversion. Intra T-lymphocyte cytokines IL-2 and TNF-alpha, lymphocyte-activation surface markers (CD25 and CD71), T-cell proliferation (PCNA+ PI(high)), total lymphocyte count, as well as lymphocytes subsets (CD2, CD3, CD4, CD8, CD19, NK cells) were measured by flow cytometry before conversion and at M1, M2, M3, and M6. RESULTS: We found no significant differences of MMF versus EC-MPS on lymphocyte function. T-cell proliferation and T-cell activation (CD25 and CD71 expression), but not cytokine expression (TNF-alpha and IL-2), showed a trend to increase after conversion from MMF to EC-MPS. Total lymphocyte, CD2, CD3, CD4, CD8, and NK cells counts were not significantly modified. CONCLUSION: This study revealed a trend to a lower immunosuppression with EC-MPS as compared to MMF in stable renal transplant patients.


Asunto(s)
Inmunosupresores/sangre , Trasplante de Riñón/inmunología , Ácido Micofenólico/análogos & derivados , Proliferación Celular/efectos de los fármacos , Química Farmacéutica , Citocinas/biosíntesis , Monitoreo de Drogas , Citometría de Flujo , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Recuento de Linfocitos , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Masculino , Persona de Mediana Edad , Ácido Micofenólico/administración & dosificación , Ácido Micofenólico/sangre , Ácido Micofenólico/farmacocinética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Comprimidos Recubiertos , Tacrolimus/administración & dosificación , Tacrolimus/farmacología
14.
Toxicology ; 246(1): 18-23, 2008 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-18055093

RESUMEN

Experimental studies of the in vivo behaviour of human cells and tissues have become possible with the development of immunodeficient mice strains. Such mice accept readily allogeneic or xenogeneic grafts, including grafts of human cells or tissues, without rejection. In this review we describe different immunodeficient mouse strains that have been used for reconstitution by human immune cells. We subsequently go through the experience that we and others have had with reconstitution, and mention the adverse effects, in particular xenogeneic graft versus host reactions. The use of haematopoietic stem cells avoids such reactions but the immunological reconstitution may take several months. We then report the use of immunodeficient mice for the study of chronic vascular rejection of human mesenteric arteries due to cellular or humoral alloreaction. We have shown that SCID/beige mice grafted with a human artery at the place of the aorta developed a thickening of the intima of the human artery after 5-6 weeks, when they were reconstituted with spleen cells from another human donor. The thickening is mainly due to a proliferation of smooth muscle cells. The same type of lesion developed if they received injection of antibodies towards HLA class I antigens. The arteries of the mouse did not develop any lesion. The arterial lesions closely resembled those seen after clinical organ transplantation. Mice that received spleen cells from the same human donor developed little or no lesions. An important aspect of this experimental transplantation model is the possibility to test drugs that may be used in clinical transplantation. In recent experiments we have shown that novel immunosuppressive drugs can inhibit the hyperproliferation of smooth muscle cells in vitro. Preclinical testing in reconstituted SCID/beige mice grafted with human arteries will permit the evaluation of the potential use of these drugs to prevent chronic vascular rejection. The model also allows pharmacodynamic studies that give information on the biological impact of different drugs that may be used in experimental or clinical transplantation.


Asunto(s)
Rechazo de Injerto/inmunología , Inmunosupresores/efectos adversos , Arterias Mesentéricas/trasplante , Modelos Biológicos , Trasplante de Células Madre , Animales , Rechazo de Injerto/prevención & control , Humanos , Transfusión de Linfocitos , Ratones , Ratones SCID , Especificidad de la Especie , Bazo/inmunología
15.
Sci Signal ; 10(492)2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811382

RESUMEN

Endothelial dysfunction, a hallmark of vascular disease, is restored by plasma high-density lipoprotein (HDL). However, a generalized increase in HDL abundance is not beneficial, suggesting that specific HDL species mediate protective effects. Apolipoprotein M-containing HDL (ApoM+HDL), which carries the bioactive lipid sphingosine 1-phosphate (S1P), promotes endothelial function by activating G protein-coupled S1P receptors. Moreover, HDL-bound S1P is limiting in several inflammatory, metabolic, and vascular diseases. We report the development of a soluble carrier for S1P, ApoM-Fc, which activated S1P receptors in a sustained manner and promoted endothelial function. In contrast, ApoM-Fc did not modulate circulating lymphocyte numbers, suggesting that it specifically activated endothelial S1P receptors. ApoM-Fc administration reduced blood pressure in hypertensive mice, attenuated myocardial damage after ischemia/reperfusion injury, and reduced brain infarct volume in the middle cerebral artery occlusion model of stroke. Our proof-of-concept study suggests that selective and sustained targeting of endothelial S1P receptors by ApoM-Fc could be a viable therapeutic strategy in vascular diseases.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Hipertensión/prevención & control , Lisofosfolípidos/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Daño por Reperfusión/prevención & control , Esfingosina/análogos & derivados , Animales , Apolipoproteínas M/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Lipoproteínas HDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Receptores Fc/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología
16.
Sci Signal ; 8(389): ra79, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26268607

RESUMEN

The sphingosine 1-phosphate receptor 1 (S1P1) is abundant in endothelial cells, where it regulates vascular development and microvascular barrier function. In investigating the role of endothelial cell S1P1 in adult mice, we found that the endothelial S1P1 signal was enhanced in regions of the arterial vasculature experiencing inflammation. The abundance of proinflammatory adhesion proteins, such as ICAM-1, was enhanced in mice with endothelial cell-specific deletion of S1pr1 and suppressed in mice with endothelial cell-specific overexpression of S1pr1, suggesting a protective function of S1P1 in vascular disease. The chaperones ApoM(+)HDL (HDL) or albumin bind to sphingosine 1-phosphate (S1P) in the circulation; therefore, we tested the effects of S1P bound to each chaperone on S1P1 signaling in cultured human umbilical vein endothelial cells (HUVECs). Exposure of HUVECs to ApoM(+)HDL-S1P, but not to albumin-S1P, promoted the formation of a cell surface S1P1-ß-arrestin 2 complex and attenuated the ability of the proinflammatory cytokine TNFα to activate NF-κB and increase ICAM-1 abundance. Although S1P bound to either chaperone induced MAPK activation, albumin-S1P triggered greater Gi activation and receptor endocytosis. Endothelial cell-specific deletion of S1pr1 in the hypercholesterolemic Apoe(-/-) mouse model of atherosclerosis enhanced atherosclerotic lesion formation in the descending aorta. We propose that the ability of ApoM(+)HDL to act as a biased agonist on S1P1 inhibits vascular inflammation, which may partially explain the cardiovascular protective functions of HDL.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Lipoproteínas HDL/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Vasculitis/metabolismo , Animales , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas M , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Lipoproteínas HDL/genética , Lisofosfolípidos/genética , Ratones , Ratones Noqueados , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/genética , Esfingosina/genética , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Vasculitis/genética , Vasculitis/patología
17.
Nat Med ; 21(9): 1028-1037, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26301690

RESUMEN

Endothelial dysfunction is a critical factor in many cardiovascular diseases, including hypertension. Although lipid signaling has been implicated in endothelial dysfunction and cardiovascular disease, specific molecular mechanisms are poorly understood. Here we report that Nogo-B, a membrane protein of the endoplasmic reticulum, regulates endothelial sphingolipid biosynthesis with direct effects on vascular function and blood pressure. Nogo-B inhibits serine palmitoyltransferase, the rate-limiting enzyme of the de novo sphingolipid biosynthetic pathway, thereby controlling production of endothelial sphingosine 1-phosphate and autocrine, G protein-coupled receptor-dependent signaling by this metabolite. Mice lacking Nogo-B either systemically or specifically in endothelial cells are hypotensive, resistant to angiotensin II-induced hypertension and have preserved endothelial function and nitric oxide release. In mice that lack Nogo-B, pharmacological inhibition of serine palmitoyltransferase with myriocin reinstates endothelial dysfunction and angiotensin II-induced hypertension. Our study identifies Nogo-B as a key inhibitor of local sphingolipid synthesis and shows that autocrine sphingolipid signaling within the endothelium is critical for vascular function and blood pressure homeostasis.


Asunto(s)
Presión Sanguínea , Endotelio Vascular/fisiología , Homeostasis , Proteínas de la Mielina/fisiología , Esfingolípidos/metabolismo , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Lisofosfolípidos/fisiología , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteínas Nogo , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Esfingosina/fisiología
18.
J Exp Med ; 209(12): 2137-40, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-23166370

RESUMEN

Recent work has highlighted the multitude of biological functions of sphingosine 1-phosphate (S1P), which include roles in hematopoietic cell trafficking, organization of immune organs, vascular development, and neuroinflammation. Indeed, a functional antagonist of S1P(1) receptor, FTY720/Gilenya, has entered the clinic as a novel therapeutic for multiple sclerosis. In this issue of the JEM, Zhang et al. highlight yet another function of this lipid mediator: thrombopoiesis. The S1P(1) receptor is required for the growth of proplatelet strings in the bloodstream and the shedding of platelets into the circulation. Notably, the sharp gradient of S1P between blood and the interstitial fluids seems to be essential to ensure the production of platelets, and S1P appears to cooperate with the CXCL12-CXCR4 axis. Pharmacologic modulation of the S1P(1) receptor altered circulating platelet numbers acutely, suggesting a potential therapeutic strategy for controlling thrombocytopenic states. However, the S1P(4) receptor may also regulate thrombopoiesis during stress-induced accelerated platelet production. This work reveals a novel physiological action of the S1P/S1P(1) duet that could potentially be harnessed for clinical translation.


Asunto(s)
Plaquetas/fisiología , Movimiento Celular/fisiología , Lisofosfolípidos/fisiología , Modelos Biológicos , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Trombopoyesis/fisiología , Plaquetas/metabolismo , Quimiocina CXCL12/metabolismo , Líquido Extracelular/metabolismo , Lisofosfolípidos/sangre , Lisofosfolípidos/metabolismo , Receptores CXCR4/metabolismo , Esfingosina/sangre , Esfingosina/metabolismo , Esfingosina/fisiología
19.
Dev Cell ; 23(3): 600-10, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22975328

RESUMEN

During angiogenesis, nascent vascular sprouts fuse to form vascular networks, enabling efficient circulation. Mechanisms that stabilize the vascular plexus are not well understood. Sphingosine 1-phosphate (S1P) is a blood-borne lipid mediator implicated in the regulation of vascular and immune systems. Here we describe a mechanism by which the G protein-coupled S1P receptor-1 (S1P1) stabilizes the primary vascular network. A gradient of S1P1 expression from the mature regions of the vascular network to the growing vascular front was observed. In the absence of endothelial S1P1, adherens junctions are destabilized, barrier function is breached, and flow is perturbed, resulting in abnormal vascular hypersprouting. Interestingly, S1P1 responds to S1P as well as laminar shear stress to transduce flow-mediated signaling in endothelial cells both in vitro and in vivo. These data demonstrate that blood flow and circulating S1P activate endothelial S1P1 to stabilize blood vessels in development and homeostasis.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Animales , Hemorreología , Homeostasis , Ratones , Receptores de Lisoesfingolípidos/sangre
20.
PLoS One ; 5(3): e9826, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20352118

RESUMEN

BACKGROUND: Sphingomyelin hydrolysis in response to stress-inducing agents, and subsequent ceramide generation, are implicated in various cellular responses, including apoptosis, inflammation and proliferation, depending on the nature of the different acidic or neutral sphingomyelinases. This study was carried out to investigate whether the neutral Mg(2+)-dependent neutral sphingomyelinase-2 (nSMase2) plays a role in the cellular signaling evoked by TNFalpha and oxidized LDLs, two stress-inducing agents, which are mitogenic at low concentrations and proapoptotic at higher concentrations. METHODOLOGY AND PRINCIPAL FINDINGS: For this purpose, we used nSMase2-deficient cells from homozygous fro/fro (fragilitas ossium) mice and nSMase2-deficient cells reconstituted with a V5-tagged nSMase2. We report that the genetic defect of nSMase2 (in fibroblasts from fro/fro mice) does not alter the TNFalpha and oxidized LDLs-mediated apoptotic response. Likewise, the hepatic toxicity of TNFalpha is similar in wild type and fro mice, thus is independent of nSMase2 activation. In contrast, the mitogenic response elicited by low concentrations of TNFalpha and oxidized LDLs (but not fetal calf serum) requires nSMase2 activation. CONCLUSION AND SIGNIFICANCE: nSMase2 activation is not involved in apoptosis mediated by TNFalpha and oxidized LDLs in murine fibroblasts, and in the hepatotoxicity of TNFalpha in mice, but is required for the mitogenic response to stress-inducing agents.


Asunto(s)
Apoptosis , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proliferación Celular , Fibroblastos/metabolismo , Vectores Genéticos , Humanos , Hidrólisis , Lipoproteínas LDL/metabolismo , Hígado/patología , Ratones , Ratones Transgénicos , Esfingomielina Fosfodiesterasa/biosíntesis , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA