Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 183(3): 771-785.e12, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125892

RESUMEN

Trained innate immunity, induced via modulation of mature myeloid cells or their bone marrow progenitors, mediates sustained increased responsiveness to secondary challenges. Here, we investigated whether anti-tumor immunity can be enhanced through induction of trained immunity. Pre-treatment of mice with ß-glucan, a fungal-derived prototypical agonist of trained immunity, resulted in diminished tumor growth. The anti-tumor effect of ß-glucan-induced trained immunity was associated with transcriptomic and epigenetic rewiring of granulopoiesis and neutrophil reprogramming toward an anti-tumor phenotype; this process required type I interferon signaling irrespective of adaptive immunity in the host. Adoptive transfer of neutrophils from ß-glucan-trained mice to naive recipients suppressed tumor growth in the latter in a ROS-dependent manner. Moreover, the anti-tumor effect of ß-glucan-induced trained granulopoiesis was transmissible by bone marrow transplantation to recipient naive mice. Our findings identify a novel and therapeutically relevant anti-tumor facet of trained immunity involving appropriate rewiring of granulopoiesis.


Asunto(s)
Granulocitos/inmunología , Inmunidad Innata , Neoplasias/inmunología , Inmunidad Adaptativa , Traslado Adoptivo , Animales , Epigénesis Genética , Interferón Tipo I/metabolismo , Ratones Endogámicos C57BL , Monocitos/metabolismo , Neoplasias/patología , Neutrófilos/metabolismo , Fenotipo , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/metabolismo , Transcripción Genética , Transcriptoma/genética , beta-Glucanos/metabolismo
2.
Cell ; 172(1-2): 135-146.e9, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29328908

RESUMEN

Innate immune cells can develop long-term memory after stimulation by microbial products during infections or vaccinations. Here, we report that metabolic signals can induce trained immunity. Pharmacological and genetic experiments reveal that activation of the cholesterol synthesis pathway, but not the synthesis of cholesterol itself, is essential for training of myeloid cells. Rather, the metabolite mevalonate is the mediator of training via activation of IGF1-R and mTOR and subsequent histone modifications in inflammatory pathways. Statins, which block mevalonate generation, prevent trained immunity induction. Furthermore, monocytes of patients with hyper immunoglobulin D syndrome (HIDS), who are mevalonate kinase deficient and accumulate mevalonate, have a constitutive trained immunity phenotype at both immunological and epigenetic levels, which could explain the attacks of sterile inflammation that these patients experience. Unraveling the role of mevalonate in trained immunity contributes to our understanding of the pathophysiology of HIDS and identifies novel therapeutic targets for clinical conditions with excessive activation of trained immunity.


Asunto(s)
Inmunidad Innata , Memoria Inmunológica , Deficiencia de Mevalonato Quinasa/inmunología , Ácido Mevalónico/metabolismo , Monocitos/inmunología , Animales , Células Cultivadas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo
3.
Cell ; 172(1-2): 147-161.e12, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29328910

RESUMEN

Trained innate immunity fosters a sustained favorable response of myeloid cells to a secondary challenge, despite their short lifespan in circulation. We thus hypothesized that trained immunity acts via modulation of hematopoietic stem and progenitor cells (HSPCs). Administration of ß-glucan (prototypical trained-immunity-inducing agonist) to mice induced expansion of progenitors of the myeloid lineage, which was associated with elevated signaling by innate immune mediators, such as IL-1ß and granulocyte-macrophage colony-stimulating factor (GM-CSF), and with adaptations in glucose metabolism and cholesterol biosynthesis. The trained-immunity-related increase in myelopoiesis resulted in a beneficial response to secondary LPS challenge and protection from chemotherapy-induced myelosuppression in mice. Therefore, modulation of myeloid progenitors in the bone marrow is an integral component of trained immunity, which to date, was considered to involve functional changes of mature myeloid cells in the periphery.


Asunto(s)
Inmunidad Innata , Memoria Inmunológica , Células Progenitoras Mieloides/inmunología , Animales , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/efectos de los fármacos , Mielopoyesis/inmunología , beta-Glucanos/farmacología
4.
Nat Immunol ; 20(1): 40-49, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30455459

RESUMEN

Resolution of inflammation is essential for tissue homeostasis and represents a promising approach to inflammatory disorders. Here we found that developmental endothelial locus-1 (DEL-1), a secreted protein that inhibits leukocyte-endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and mouse periodontitis, waning of inflammation was correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium-urate-crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver X receptor-dependent macrophage reprogramming to a pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte-recruitment action to endothelial cell-derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamación/inmunología , Macrófagos/fisiología , Neutrófilos/inmunología , Periodontitis/inmunología , Adulto , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/genética , Moléculas de Adhesión Celular , Reprogramación Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/inducido químicamente , Péptidos y Proteínas de Señalización Intercelular , Células K562 , Receptores X del Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis
5.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34663697

RESUMEN

Trained immunity defines long-lasting adaptations of innate immunity based on transcriptional and epigenetic modifications of myeloid cells and their bone marrow progenitors [M. Divangahi et al., Nat. Immunol. 22, 2-6 (2021)]. Innate immune cells, however, do not exclusively differentiate between foreign and self but also react to host-derived molecules referred to as alarmins. Extracellular "labile" heme, released during infections, is a bona fide alarmin promoting myeloid cell activation [M. P. Soares, M. T. Bozza, Curr. Opin. Immunol. 38, 94-100 (2016)]. Here, we report that labile heme is a previously unrecognized inducer of trained immunity that confers long-term regulation of lineage specification of hematopoietic stem cells and progenitor cells. In contrast to previous reports on trained immunity, essentially mediated by pathogen-associated molecular patterns, heme training depends on spleen tyrosine kinase signal transduction pathway acting upstream of c-Jun N-terminal kinases. Heme training promotes resistance to sepsis, is associated with the expansion of self-renewing hematopoetic stem cells primed toward myelopoiesis and to the occurrence of a specific myeloid cell population. This is potentially evoked by sustained activity of Nfix, Runx1, and Nfe2l2 and dissociation of the transcriptional repressor Bach2. Previously reported trained immunity inducers are, however, infrequently present in the host, whereas heme abundantly occurs during noninfectious and infectious disease. This difference might explain the vanishing protection exerted by heme training in sepsis over time with sustained long-term myeloid adaptations. Hence, we propose that trained immunity is an integral component of innate immunity with distinct functional differences on infectious disease outcome depending on its induction by pathogenic or endogenous molecules.


Asunto(s)
Epigénesis Genética , Hemo/fisiología , Inmunidad Innata , Mielopoyesis , Animales , Humanos , Ratones
6.
Hepatology ; 75(4): 881-897, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34519101

RESUMEN

BACKGROUND AND AIMS: NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS: Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS: HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.


Asunto(s)
Carcinoma Hepatocelular , Proteína 1 Similar a ELAV , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Animales , Carcinoma Hepatocelular/patología , Proteína 1 Similar a ELAV/metabolismo , Homeostasis , Inflamación/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología , ARN , Triglicéridos/metabolismo
7.
Blood ; 137(24): 3416-3427, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33619535

RESUMEN

Orchestrated recruitment of neutrophils to inflamed tissue is essential during the initiation of inflammation. Inflamed areas are usually hypoxic, and adaptation to reduced oxygen pressure is typically mediated by hypoxia pathway proteins. However, it remains unclear how these factors influence the migration of neutrophils to and at the site of inflammation during their transmigration through the blood-endothelial cell barrier, as well as their motility in the interstitial space. Here, we reveal that activation of hypoxia-inducible factor 2 (HIF2α) as a result of a deficiency in HIF prolyl hydroxylase domain protein 2 (PHD2) boosts neutrophil migration specifically through highly confined microenvironments. In vivo, the increased migratory capacity of PHD2-deficient neutrophils resulted in massive tissue accumulation in models of acute local inflammation. Using systematic RNA sequencing analyses and mechanistic approaches, we identified RhoA, a cytoskeleton organizer, as the central downstream factor that mediates HIF2α-dependent neutrophil motility. Thus, we propose that the novel PHD2-HIF2α-RhoA axis is vital to the initial stages of inflammation because it promotes neutrophil movement through highly confined tissue landscapes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Movimiento Celular , Microambiente Celular , Neutrófilos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Inflamación/genética , Inflamación/metabolismo , Ratones , Ratones Noqueados , RNA-Seq
8.
FASEB J ; 34(2): 3336-3346, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31916652

RESUMEN

In Type 1 Diabetes Mellitus (T1DM), leukocyte infiltration of the pancreatic islets and the resulting immune-mediated destruction of beta cells precede hyperglycemia and clinical disease symptoms. In this context, the role of the pancreatic endothelium as a barrier for autoimmunity- and inflammation-related destruction of the islets is not well studied. Here, we identified Robo4, expressed on endothelial cells, as a regulator of pancreatic vascular endothelial permeability during autoimmune diabetes. Circulating levels of Robo4 were upregulated in mice subjected to the Multiple Low-Dose Streptozotocin (MLDS) model of diabetes. Upon MLDS induction, Robo4-deficiency resulted in increased pancreatic vascular permeability, leukocyte infiltration to the islets and islet apoptosis, associated with reduced insulin levels and faster diabetes development. On the contrary, in vivo administration of Slit2 in mice modestly delayed the emergence of hyperglycaemia and ameliorated islet inflammation in MLDS-induced diabetes. Thus, Robo4-mediated endothelial barrier integrity reduces insulitis and islet destruction in autoimmune diabetes. Our findings highlight the importance of the endothelium as gatekeeper of pancreatic inflammation during T1DM development and may pave the way for novel Robo4-related therapeutic approaches for autoimmune diabetes.


Asunto(s)
Permeabilidad Capilar , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Endoteliales/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Apoptosis , Línea Celular , Células Cultivadas , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Células Endoteliales/patología , Humanos , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos C57BL , Receptores de Superficie Celular/sangre , Receptores de Superficie Celular/genética
9.
Xenotransplantation ; 24(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27677785

RESUMEN

BACKGROUND: The complement system plays a crucial role in acute xenogeneic reactions after cardiac transplantation. We used an ex vivo perfusion model to investigate the effect of Cp40, a compstatin analog and potent inhibitor of complement at the level of C3. METHODS: Fifteen wild-type pig hearts were explanted, cardiopleged, and reperfused ex vivo after 150 minutes of cold ischemia. Hearts were challenged in a biventricular working heart mode to evaluate cardiac perfusion and function. In the treatment group (n=5), the complement cascade was blocked at the level of C3 using Cp40, using diluted human blood. Untreated human and porcine blood was used for controls. RESULTS: Throughout the perfusion, C3 activation was inhibited when Cp40 was used (mean of all time points: 1.11 ± 0.34% vs 3.12 ± 0.48% control activation; P<.01). Compared to xenoperfused controls, the cardiac index improved significantly in the treated group (6.5 ± 4.2 vs 3.5 ± 4.8 mL/min/g; P=.03, 180 minutes perfusion), while the concentration of lactate dehydrogenase as a maker for cell degradation was reduced in the perfusate (583 ± 187 U/mL vs 2108 ± 1145 U/mL, P=.02). Histological examination revealed less hemorrhage and edema, and immunohistochemistry confirmed less complement fragment deposition than in untreated xenoperfused controls. CONCLUSIONS: Cp40 efficiently prevents C3 activation of the complement system, resulting in reduced cell damage and preserved function in wild-type porcine hearts xenoperfused ex vivo. We suggest that this compstatin analog, which blocks all main pathways of complement activation, could be a beneficial perioperative treatment in preclinical and in future clinical xenotransplantation.


Asunto(s)
Activación de Complemento/inmunología , Complemento C3/metabolismo , Trasplante de Corazón , Piridonas/metabolismo , Animales , Rechazo de Injerto/prevención & control , Corazón , Trasplante de Corazón/métodos , Humanos , Miocardio/inmunología , Porcinos , Trasplante Heterólogo/métodos
10.
J Immunol ; 194(3): 1285-91, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25548229

RESUMEN

Wound healing is a complex homeostatic response to injury that engages numerous cellular activities, processes, and cell-to-cell interactions. The complement system, an intricate network of proteins with important roles in immune surveillance and homeostasis, has been implicated in many physiological processes; however, its role in wound healing remains largely unexplored. In this study, we employ a murine model of excisional cutaneous wound healing and show that C3(-/-) mice exhibit accelerated early stages of wound healing. Reconstitution of C3(-/-) mice with serum from C3(+/+) mice or purified human C3 abrogated the accelerated wound-healing phenotype. Wound histology of C3(-/-) mice revealed a reduction in inflammatory infiltrate compared with C3(+/+) mice. C3 deficiency also resulted in increased accumulation of mast cells and advanced angiogenesis. We further show that mice deficient in the downstream complement effector C5 exhibit a similar wound-healing phenotype, which is recapitulated in C5aR1(-/-) mice, but not C3aR(-/-) or C5aR2(-/-) mice. Taken together, these data suggest that C5a signaling through C5aR may in part play a pivotal role in recruitment and activation of inflammatory cells to the wound environment, which in turn could delay the early stages of cutaneous wound healing. These findings also suggest a previously underappreciated role for complement in wound healing, and may have therapeutic implications for conditions of delayed wound healing.


Asunto(s)
Proteínas del Sistema Complemento/deficiencia , Piel/inmunología , Piel/lesiones , Cicatrización de Heridas/inmunología , Animales , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/inmunología , Complemento C5a/genética , Complemento C5a/inmunología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Modelos Inmunológicos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/genética
11.
Adv Exp Med Biol ; 865: 171-88, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26306450

RESUMEN

Xeno-transplantation of pancreatic islets represents a promising therapeutic alternative for the treatment of type 1 diabetes mellitus. However, potent innate immune responses induced shortly after the transplantation of donor islets to the recipient, comprising the Instant Blood Mediated Immune Reaction (IBMIR), exert detrimental actions on islet graft function. The coagulation and complement cascades together with the leukocyte and platelet populations are the major players in IBMIR. This innate immune attack affects dramatically islet integrity and leads to significant loss of function of the xenograft. In the present review, we focus on the mechanisms contributing to IBMIR components and address therapeutic intervention approaches to limit IBMIR by administering inhibitors in circulation, by coating the islet surface with inhibitors or by generating transgenic donor animals; these approaches could result in improved xenograft survival.


Asunto(s)
Diabetes Mellitus/terapia , Rechazo de Injerto/prevención & control , Trasplante de Islotes Pancreáticos/métodos , Transgenes , Animales , Animales Modificados Genéticamente , Antígenos CD/genética , Antígenos CD/inmunología , Factores de Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/inmunología , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/patología , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Sulfato de Dextran/farmacología , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Diabetes Mellitus/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/patología , Péptidos Cíclicos/farmacología , Porcinos , Trasplante Heterólogo
12.
FASEB J ; 27(7): 2768-76, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23558338

RESUMEN

Although complement is a known contributor to biomaterial-induced complications, pathological implications and therapeutic options remain to be explored. Here we investigated the involvement of complement in the inflammatory response to polypropylene meshes commonly used for hernia repair. In vitro assays revealed deposition of complement activation fragments on the mesh after incubation in plasma. Moreover, significant mesh-induced complement and granulocyte activation was observed in plasma and leukocyte preparations, respectively. Pretreatment of plasma with the complement inhibitor compstatin reduced opsonization >2-fold, and compstatin and a C5a receptor antagonist (C5aRa) impaired granulocyte activation by 50 and 67%, respectively. We established a clinically relevant mouse model of implantation and could confirm deposition of C3 activation fragments on mesh implants in vivo using immunofluorescence. In meshes extracted after subcutaneous or peritoneal implantation, the amount of immune cell infiltrate in mice deficient in key complement components (C3, C5aR), or treated with C5aRa, was approximately half of that observed in wild-type littermates or mice treated with inactive C5aRa, respectively. Our data suggest that implantation of a widely used surgical mesh triggers the formation of an inflammatory cell microenvironment at the implant site through complement activation, and indicates a path for the therapeutic modulation of implant-related complications.


Asunto(s)
Materiales Biocompatibles/farmacología , Activación de Complemento/efectos de los fármacos , Inflamación/prevención & control , Polipropilenos/farmacología , Animales , Antígeno CD11b/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Citometría de Flujo , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Humanos , Implantes Experimentales/efectos adversos , Inflamación/etiología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Proteínas Opsoninas/metabolismo , Péptidos Cíclicos/farmacología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Quimiocina/metabolismo
13.
J Immunol ; 188(2): 641-8, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22184721

RESUMEN

The involvement of IL-4 in liver regeneration has not yet been recognized. In this article, we show that IL-4, produced by NKT cells that accumulate in regenerating livers after partial hepatectomy, contributes to this process by regulating the activation of complement after liver resection in mice. The mechanism of this regulation was associated with the maintenance of an appropriate level of IgM in mouse blood, because IgM deposited in liver parenchyma most likely initiated complement activation during liver regeneration. By controlling complement activation, IL-4 regulated the induction of IL-6, thereby influencing a key pathway involved in regenerating liver cell proliferation and survival. Furthermore, the secretion of IL-4 was controlled by complement through the recruitment of NKT cells to regenerating livers. Our study thus reveals the existence of a regulatory feedback mechanism involving complement and IL-4 that controls liver regeneration.


Asunto(s)
Complemento C3/fisiología , Interleucina-4/fisiología , Regeneración Hepática/inmunología , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C3/deficiencia , Citocinas/biosíntesis , Hepatectomía , Hepatocitos/citología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Inmunoglobulina M/sangre , Interleucina-4/biosíntesis , Interleucina-4/deficiencia , Regeneración Hepática/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo
14.
J Immunol ; 186(11): 6568-75, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531894

RESUMEN

Neonatal respiratory distress syndrome can progress to bronchopulmonary dysplasia (BPD), a serious pulmonary fibrotic disorder. Given the involvement of the extrinsic coagulation cascade in animal models of lung fibrosis, we examined its role in BPD. We observed a higher number of neutrophils expressing tissue factor (TF) in bronchoalveolar lavage fluid (BALF) from infants with BPD than from those with uncomplicated respiratory distress syndrome together with a parallel decrease in TF and connective tissue growth factor (CTGF) in BALF supernatants during the disease course. The involvement of coagulation in the fibrotic process associated with BPD was further evaluated by treating primary human colonic myofibroblasts with BALF supernatants from infants with BPD. These human colonic myofibroblasts demonstrated an enhanced C5a- and thrombin-dependent migration. Moreover, they expressed TF in an endothelin-1-dependent manner, with subsequent activation of the extrinsic coagulation cascade and CTGF production mediated by protease-activator receptor-1 signaling. These data provide a novel mechanism for the development of BPD and indicate that endothelin-1 signaling contributes to fibrosis by upregulating a TF/thrombin amplification loop responsible for CTGF production, and offer novel and specific therapeutic targets for pulmonary fibrotic disease.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Endotelina-1/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/metabolismo , Transducción de Señal , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patología , Células Cultivadas , Colon/metabolismo , Colon/patología , Complemento C5a/genética , Complemento C5a/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Endotelina-1/genética , Femenino , Fibrosis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Recién Nacido , Pulmón/metabolismo , Pulmón/patología , Masculino , Microscopía Fluorescente , Miofibroblastos/metabolismo , Miofibroblastos/patología , Receptor de Anafilatoxina C5a , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombina/genética , Trombina/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo
15.
Blood ; 116(4): 631-9, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20424189

RESUMEN

Thrombosis is a common complication of end-stage renal disease, particularly in patients on hemodialysis. Although substantial progress has been made in preventing thrombotic complications in various other groups of patients, the mechanisms of thrombosis during hemodialysis require clarification. In this report, we demonstrate that complement activation triggered by hemodialysis biomaterials, and the subsequent generation of the complement anaphylatoxin C5a, results in the expression of functionally active tissue factor (TF) in peripheral blood neutrophils. Because TF is a key initiator of coagulation in vivo, we postulate that the recurring complement activation that occurs during long-term hemodialysis contributes to thrombosis in dialyzed end-stage renal disease patients. Furthermore, we found that complement contributed to the induction of granulocyte colony-stimulating factor, which has been implicated in the pathogenesis of thrombosis in patients treated with the recombinant form of this molecule. Importantly, the inhibition of complement activation attenuated the TF expression and granulocyte colony-stimulating factor induction in blood passing through a hemodialysis circuit, suggesting that the complement system could become a new therapeutic target for preventing thrombosis in patients with chronic renal failure who are maintained on hemodialysis.


Asunto(s)
Complemento C5a/fisiología , Diálisis Renal/efectos adversos , Trombosis/etiología , Anciano , Anafilatoxinas/farmacología , Anafilatoxinas/fisiología , Coagulación Sanguínea/efectos de los fármacos , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Complemento C5a/metabolismo , Complemento C5a/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/terapia , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Persona de Mediana Edad , Receptor de Anafilatoxina C5a/metabolismo , Tromboplastina/metabolismo , Factores de Tiempo
16.
Microb Cell Fact ; 11: 162, 2012 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-23259547

RESUMEN

BACKGROUND: Terpenoids and isoprenoids are an important class of natural products, which includes currently used drugs, high value bioactive and industrial compounds, and fuel candidates. Due to their industrial application, there is increasing interest in the development of S. cerevisiae strains capable of producing high levels of terpenoids. RESULTS: Aiming to identify new gene targets which can be manipulated to increase sesquiterpene production, a set of HMG2 positive genetic interactors were assessed as single and digenic heterozygous deletions in the presence or absence of stable HMG2(K6R) overexpression. Upon single allele deletion, most genes examined led to increased sesquiterpene production in yeast cells. Tandem heterozygous deletion of a set of three genes, the ubiquitin ligases ubc7 and ssm4/doa10, and the ER resident protein pho86, led to an 11-fold increase in caryophyllene yields (125 mg/L in shake flasks) compared to cells lacking these modifications. The effect of the heterozygous deletions appears to be due to Hmg1p and Hmg2p stabilization. CONCLUSION: Heterozygous deletions cause significant reductions in protein levels but do not lead to growth impediments frequently seen in haploid strains. By exploiting desirable haploinsufficiencies in yeast, we identified a new set of genes that can be disrupted in tandem and cause significant stabilization of Hmgp and a substantial increase in sesquiterpene production. The approach presented here allows new genetic perturbations to be compiled on yeast cell factory strains without negatively impacting cell growth and viability.


Asunto(s)
Proteína HMGB2/genética , Saccharomyces cerevisiae/metabolismo , Sesquiterpenos/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Alelos , Eliminación de Gen , Proteína HMGB1/genética , Sesquiterpenos Policíclicos , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
17.
Arthritis Rheum ; 63(11): 3586-97, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21834070

RESUMEN

OBJECTIVE: The extrinsic coagulation cascade is involved in the fibrotic process, via thrombin-dependent induction of CCN2 (connective tissue growth factor) expression. Given the previously reported activation of the coagulation system in systemic sclerosis (SSc), we undertook the present study to investigate the involvement of cross-talk between the tissue factor (TF)-thrombin axis and endothelin 1 (ET-1) signaling in the fibrotic activity of SSc. METHODS: Human colonic myofibroblasts (HCMFs) from 6 patients with SSc and gastrointestinal symptoms and from 6 control subjects were isolated and cultured under various conditions. Messenger RNA and protein levels of TF, CCN2, and endothelin receptor A (ET(A) ) were investigated. Collagen production and migratory activity of HCMFs were further assessed. RESULTS: HCMFs from SSc patients demonstrated increased basal CCN2 production, collagen deposition, and migration rate, in a thrombin-dependent manner. Increased TF expression was also observed in SSc HCMFs. Subsequent activation of the extrinsic coagulation system resulted in thrombin-dependent enhancement of ET(A) expression. ET(A) overexpression led to further increases in both TF expression and fibrotic activity in HCMFs. Moreover, inhibition of ET-1 signaling by bosentan abolished the TF-mediated fibrotic capacity of HCMFs. CONCLUSION: Tissue factor-thrombin signaling is involved in the increased fibrotic activity of HCMFs from patients with SSc. Moreover, the up-regulation of ET(A) expression by thrombin and the effect of ET-1 in the induction of TF expression indicate an amplification loop for enhanced collagen deposition. Therapeutic interventions targeting the extrinsic coagulation system or ET-1 signaling may provide clinical benefit by breaking this vicious circle.


Asunto(s)
Colon/metabolismo , Miofibroblastos/metabolismo , Receptor de Endotelina A/metabolismo , Esclerodermia Sistémica/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo , Movimiento Celular , Proliferación Celular , Colágeno Tipo I/metabolismo , Colon/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibrosis , Humanos , Miofibroblastos/patología , Receptor PAR-1/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal , Regulación hacia Arriba
18.
Mol Biol Rep ; 39(5): 5503-10, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22187344

RESUMEN

Familial Mediterranean fever (FMF) is an autoinflammatory disease, characterized by MEFV gene mutations and self-limited recurrent episodes of fever and localized serositis. Complement system is a key regulator of the inflammatory process. The aim of this study was to investigate the genetic alterations and mRNA expression pattern of C5aR and C5L2 genes in neutrophils from attack-free FMF patients. No mutations were observed in the two receptors' genes, while the genetic alteration observed in the C5aR1 gene was identified as N279 K polymorphic variant. Furthermore, lower mRNA expression of C5L2 gene was observed in neutrophils from FMF patients compared to control subjects. The binding capacity of rhC5a and the ability to produce reactive oxygen species was similar in neutrophils from healthy subjects and FMF patients and independent of the presence of N279 K polymorphism or mRNA expression of C5L2.


Asunto(s)
Fiebre Mediterránea Familiar/genética , Fiebre Mediterránea Familiar/patología , Predisposición Genética a la Enfermedad , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Quimiocina/genética , Receptores de Complemento/genética , Adolescente , Adulto , Secuencia de Bases , Niño , Preescolar , Proteínas del Citoesqueleto/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación/genética , Polimorfismo de Longitud del Fragmento de Restricción/genética , Polimorfismo de Nucleótido Simple/genética , Pirina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a
19.
J Clin Invest ; 132(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35133978

RESUMEN

The causative role of inflammation in hypertension-related cardiovascular diseases is evident and calls for development of specific immunomodulatory therapies. We tested the therapeutic efficacy and mechanisms of action of developmental endothelial locus-1 (DEL-1), an endogenous antiinflammatory factor, in angiotensin II- (ANGII-) and deoxycorticosterone acetate-salt-induced (DOCA-salt-induced) cardiovascular organ damage and hypertension. By using mice with endothelial overexpression of DEL-1 (EC-Del1 mice) and performing preventive and interventional studies by injecting recombinant DEL-1 in mice, we showed that DEL-1 improved endothelial function and abrogated aortic adventitial fibrosis, medial thickening, and loss of elastin. DEL-1 also protected the mice from cardiac concentric hypertrophy and interstitial and perivascular coronary fibrosis and improved left ventricular function and myocardial coronary perfusion. DEL-1 prevented aortic stiffness and abolished the progression of hypertension. Mechanistically, DEL-1 acted by inhibiting αvß3 integrin-dependent activation of pro-MMP2 in mice and in human isolated aorta. Moreover, DEL-1 stabilized αvß3 integrin-dependent CD25+FoxP3+ Treg numbers and IL-10 levels, which were associated with decreased recruitment of inflammatory cells and reduced production of proinflammatory cytokines in cardiovascular organs. The demonstrated effects and immune-modulating mechanisms of DEL-1 in abrogation of cardiovascular remodeling and progression of hypertension identify DEL-1 as a potential therapeutic factor.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , Hipertensión , Remodelación Ventricular , Animales , Cardiomegalia , Fibrosis , Hipertensión/complicaciones , Inmunomodulación/genética , Integrinas , Ratones , Remodelación Ventricular/genética
20.
Eur J Immunol ; 40(5): 1461-72, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20162553

RESUMEN

The induction of the autophagy machinery, a process for the catabolism of cytosolic proteins and organelles, constitutes a crucial mechanism in innate immunity. However, the involvement of autophagy in human neutrophils and the possible inducers of this process have not been completely elucidated. In this study, the induction of autophagy was examined in human neutrophils treated with various activators and detected by the formation of acidified autophagosomes through monodansylcadaverine staining and via LC-3B conversion screened by immunoblotting and immunofluorescence confocal microscopy. In addition, the expression of the ATG genes was assessed by real-time RT-PCR. We provide evidence that autophagy is implicated in human neutrophils in both a phagocytosis-independent (rapamycin, TLR agonists, PMA) and phagocytosis (Escherichia coli)-dependent initiation manner. ROS activation is a positive mechanism for autophagy induction in the case of PMA, TLR activation and phagocytosis. Furthermore, LC3B gene expression was uniformly upregulated, indicating a transcriptional level of regulation for the autophagic machinery. This study provides a stepping stone toward further investigation of autophagy in neutrophil-driven inflammatory disorders.


Asunto(s)
Autofagia/fisiología , Neutrófilos/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Autofagia/efectos de los fármacos , Autofagia/genética , Cadaverina/análogos & derivados , Cadaverina/análisis , Cromonas/farmacología , Colorantes/análisis , Escherichia coli , Guanosina/análogos & derivados , Guanosina/farmacología , Humanos , Concentración de Iones de Hidrógeno , Inflamación/inmunología , Microscopía Confocal , Morfolinas/farmacología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Fagosomas/fisiología , Fagosomas/ultraestructura , Poli I-C/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/biosíntesis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Acetato de Tetradecanoilforbol/farmacología , Receptores Toll-Like/efectos de los fármacos , Receptores Toll-Like/fisiología , Transcripción Genética , Vacuolas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA