Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 18(12): e0295408, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38055674

RESUMEN

AIMS: IgE type immunoglobulins and their specific effector cells, mast cells (MCs), are associated with abdominal aortic aneurysm (AAA) progression. In parallel, immunoglobulin-producing B cells, organised in tertiary lymphoid organs (TLOs) within the aortic wall, have also been linked to aneurysmal progression. We aimed at investigating the potential role and mechanism linking local MCs, TLO B cells, and IgE production in aneurysmal progression. METHODS AND RESULTS: Through histological assays conducted on human surgical samples from AAA patients, we uncovered that activated MCs were enriched at sites of unhealed haematomas, due to subclinical aortic wall fissuring, in close proximity to adventitial IgE+ TLO B cells. Remarkably, in vitro the IgEs deriving from these samples enhanced MC production of IL-4, a cytokine which favors IgE class-switching and production by B cells. Finally, the role of MCs in aneurysmal progression was further analysed in vivo in ApoE-/- mice subjected to angiotensin II infusion aneurysm model, through MC-specific depletion after the establishment of dissecting aneurysms. MC-specific depletion improved intramural haematoma healing and reduced aneurysmal progression. CONCLUSIONS: Our data suggest that MC located close to aortic wall fissures are activated by adventitial TLO B cell-produced IgEs and participate to their own activation by providing support for further IgE synthesis through IL-4 production. By preventing prompt repair of aortic subclinical fissures, such a runaway MC activation loop could precipitate aneurysmal progression, suggesting that MC-targeting treatments may represent an interesting adjunctive therapy for reducing AAA progression.


Asunto(s)
Aneurisma de la Aorta Abdominal , Mastocitos , Humanos , Ratones , Animales , Mastocitos/metabolismo , Interleucina-4/metabolismo , Ratones Noqueados para ApoE , Aneurisma de la Aorta Abdominal/patología , Inmunoglobulina E/metabolismo , Modelos Animales de Enfermedad , Aorta Abdominal/patología , Angiotensina II/metabolismo , Ratones Endogámicos C57BL
2.
Nature ; 620(7974): 643-650, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37437602

RESUMEN

In addition to its canonical function of protection from pathogens, the immune system can also alter behaviour1,2. The scope and mechanisms of behavioural modifications by the immune system are not yet well understood. Here, using mouse models of food allergy, we show that allergic sensitization drives antigen-specific avoidance behaviour. Allergen ingestion activates brain areas involved in the response to aversive stimuli, including the nucleus of tractus solitarius, parabrachial nucleus and central amygdala. Allergen avoidance requires immunoglobulin E (IgE) antibodies and mast cells but precedes the development of gut allergic inflammation. The ability of allergen-specific IgE and mast cells to promote avoidance requires cysteinyl leukotrienes and growth and differentiation factor 15. Finally, a comparison of C57BL/6 and BALB/c mouse strains revealed a strong effect of the genetic background on the avoidance behaviour. These findings thus point to antigen-specific behavioural modifications that probably evolved to promote niche selection to avoid unfavourable environments.


Asunto(s)
Alérgenos , Reacción de Prevención , Hipersensibilidad a los Alimentos , Animales , Ratones , Alérgenos/inmunología , Reacción de Prevención/fisiología , Núcleo Amigdalino Central/fisiología , Modelos Animales de Enfermedad , Hipersensibilidad a los Alimentos/genética , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Intestinos/inmunología , Mastocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Núcleos Parabraquiales/fisiología , Núcleo Solitario/fisiología
3.
bioRxiv ; 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36712030

RESUMEN

In addition to its canonical function in protecting from pathogens, the immune system can also promote behavioural alterations 1â€"3 . The scope and mechanisms of behavioural modifications by the immune system are not yet well understood. Using a mouse food allergy model, here we show that allergic sensitization drives antigen-specific behavioural aversion. Allergen ingestion activates brain areas involved in the response to aversive stimuli, including the nucleus of tractus solitarius, parabrachial nucleus, and central amygdala. Food aversion requires IgE antibodies and mast cells but precedes the development of gut allergic inflammation. The ability of allergen-specific IgE and mast cells to promote aversion requires leukotrienes and growth and differentiation factor 15 (GDF15). In addition to allergen-induced aversion, we find that lipopolysaccharide-induced inflammation also resulted in IgE-dependent aversive behaviour. These findings thus point to antigen-specific behavioural modifications that likely evolved to promote niche selection to avoid unfavourable environments.

4.
Cells ; 11(19)2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36231017

RESUMEN

Mast cells (MCs) are well-known for their role in IgE-mediated cutaneous anaphylactic responses, but their regulatory functions in the skin are still under intense scrutiny. Using a Red MC and Basophil reporter (RMB) mouse allowing red fluorescent detection and diphtheria toxin mediated depletion of MCs, we investigated the interaction of MCs, Foxp3+ regulatory T lymphocytes (Tregs) and Langerhans cells (LCs) during passive cutaneous anaphylaxis (PCA) responses. Using intravital imaging we show that MCs are sessile at homeostasis and during PCA. Breeding RMB mice with Langerin-eGFP mice revealed that dermal MCs do not interact with epidermal-localized LCs, the latter showing constant sprouting of their dendrites at homeostasis and during PCA. When bred with Foxp3-eGFP mice, we found that, although a few Foxp3+ Tregs are present at homeostasis, many Tregs transiently infiltrated the skin during PCA. While their velocity during PCA was not altered, Tregs increased the duration of their contact time with MCs compared to PCA-control mice. Antibody-mediated depletion of Tregs had no effect on the intensity of PCA. Hence, the observed increase in Treg numbers and contact time with MCs, regardless of an effect on the intensity of PCA responses, suggests an anti-inflammatory role dedicated to prevent further MC activation.


Asunto(s)
Anafilaxia , Mastocitos , Animales , Antiinflamatorios , Comunicación Celular , Dermis , Toxina Diftérica , Factores de Transcripción Forkhead , Inmunoglobulina E , Ratones , Linfocitos T Reguladores
5.
Eur J Immunol ; 51(2): 445-458, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32920851

RESUMEN

B lymphocytes are among the cell types whose effector functions are modulated by mast cells (MCs). The B/MC crosstalk emerged in several pathological settings, notably the colon of inflammatory bowel disease (IBD) patients is a privileged site in which MCs and IgA+ cells physically interact. Herein, by inducing conditional depletion of MCs in red MC and basophil (RMB) mice, we show that MCs control B cell distribution in the gut and IgA serum levels. Moreover, in dextran sulfate sodium (DSS)-treated RMB mice, the presence of MCs is fundamental for the enlargement of the IgA+ population in the bowel and the increase of systemic IgA production. Since both conventional B-2 and peritoneal-derived B cells populate the intestine and communicate with MCs in physiological conditions and during inflammation, we further explored this interplay through the use of co-cultures. We show that MCs finely regulate different aspects of splenic B cell biology while peritoneal B cells are unresponsive to the supporting effects provided by MCs. Interestingly, peritoneal B cells induce a pro-inflammatory skewing in MCs, characterized by increased ST2 and TNF-α expression. Altogether, this study uncovers the versatility of the B/MC liaison and highlights key aspects for the resolution of intestinal inflammation.


Asunto(s)
Linfocitos B/metabolismo , Colon/inmunología , Inmunoglobulina A/inmunología , Mucosa Intestinal/inmunología , Mastocitos/inmunología , Animales , Colitis/inmunología , Colon/microbiología , Sulfato de Dextran/inmunología , Microbioma Gastrointestinal/inmunología , Inflamación/inmunología , Inflamación/microbiología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/inmunología
6.
Nat Commun ; 11(1): 4786, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32963227

RESUMEN

Evidence points to an indispensable function of macrophages in tissue regeneration, yet the underlying molecular mechanisms remain elusive. Here we demonstrate a protective function for the IL-33-ST2 axis in bronchial epithelial repair, and implicate ST2 in myeloid cell differentiation. ST2 deficiency in mice leads to reduced lung myeloid cell infiltration, abnormal alternatively activated macrophage (AAM) function, and impaired epithelial repair post naphthalene-induced injury. Reconstitution of wild type (WT) AAMs to ST2-deficient mice completely restores bronchial re-epithelialization. Central to this mechanism is the direct effect of IL-33-ST2 signaling on monocyte/macrophage differentiation, self-renewal and repairing ability, as evidenced by the downregulation of key pathways regulating myeloid cell cycle, maturation and regenerative function of the epithelial niche in ST2-/- mice. Thus, the IL-33-ST2 axis controls epithelial niche regeneration by activating a large multi-cellular circuit, including monocyte differentiation into competent repairing AAMs, as well as group-2 innate lymphoid cell (ILC2)-mediated AAM activation.


Asunto(s)
Bronquiolos/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Interleucina-33/farmacología , Animales , Bronquiolos/lesiones , Bronquiolos/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Proteína 1 Similar al Receptor de Interleucina-1/genética , Pulmón/patología , Activación de Linfocitos , Linfocitos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
7.
Sci Adv ; 6(23): eabb2236, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32537514

RESUMEN

Our ability to predict the impact of mutations on traits relevant for disease and evolution remains severely limited by the dependence of their effects on the genetic background and environment. Even when molecular interactions between genes are known, it is unclear how these translate to organism-level interactions between alleles. We therefore characterized the interplay of genetic and environmental dependencies in determining fitness by quantifying ~4000 fitness interactions between expression variants of two metabolic genes, starting from various environmentally modulated expression levels. We detect a remarkable variety of interactions dependent on initial expression levels and demonstrate that they can be quantitatively explained by a mechanistic model accounting for catabolic flux, metabolite toxicity, and expression costs. Complex fitness interactions between mutations can therefore be predicted simply from their simultaneous impact on a few connected molecular phenotypes.

8.
Front Immunol ; 9: 2690, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30515167

RESUMEN

Recent evidences indicate an important role of tissue inflammatory responses by innate immune cells in allograft acceptance and survival. Here we investigated the role of mast cells (MC) in an acute male to female skin allograft rejection model using red MC and basophil (RMB) mice enabling conditional MC depletion. Kinetic analysis showed that MCs markedly accelerate skin rejection. They induced an early inflammatory response through degranulation and boosted local synthesis of KC, MIP-2, and TNF. This enhanced early neutrophil infiltration compared to a female-female graft-associated repair response. The uncontrolled neutrophil influx accelerated rejection as antibody-mediated depletion of neutrophils delayed skin rejection. Administration of cromolyn, a MC stabilizer and to a lesser extent ketotifen, a histamine type I receptor antagonist, and absence of MCPT4 chymase also delayed graft rejection. Together our data indicate that mediators contained in secretory granules of MC promote an inflammatory response with enhanced neutrophil infiltration that accelerate graft rejection.


Asunto(s)
Degranulación de la Célula/inmunología , Citocinas/inmunología , Rechazo de Injerto/inmunología , Mastocitos/inmunología , Infiltración Neutrófila , Neutrófilos/inmunología , Trasplante de Piel , Animales , Citocinas/genética , Rechazo de Injerto/genética , Rechazo de Injerto/patología , Mastocitos/patología , Ratones , Ratones Noqueados
9.
Immunity ; 48(6): 1160-1171.e5, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29858009

RESUMEN

Hematopoiesis occurs in distinct waves. "Definitive" hematopoietic stem cells (HSCs) with the potential for all blood lineages emerge in the aorta-gonado-mesonephros, while "primitive" progenitors, whose potential is thought to be limited to erythrocytes, megakaryocytes, and macrophages, arise earlier in the yolk sac (YS). Here, we questioned whether other YS lineages exist that have not been identified, partially owing to limitations of current lineage tracing models. We established the use of Cdh5-CreERT2 for hematopoietic fate mapping, which revealed the YS origin of mast cells (MCs). YS-derived MCs were replaced by definitive MCs, which maintained themselves independently from the bone marrow in the adult. Replacement occurred with tissue-specific kinetics. MCs in the embryonic skin, but not other organs, remained largely YS derived prenatally and were phenotypically and transcriptomically distinct from definite adult MCs. We conclude that within myeloid lineages, dual hematopoietic origin is shared between macrophages and MCs.


Asunto(s)
Linaje de la Célula/inmunología , Hematopoyesis/fisiología , Mastocitos/citología , Animales , Hemangioblastos/citología , Células Madre Hematopoyéticas/citología , Macrófagos/citología , Macrófagos/inmunología , Mastocitos/inmunología , Ratones , Piel/citología , Piel/inmunología , Saco Vitelino/citología , Saco Vitelino/embriología
10.
J Immunol ; 198(6): 2374-2382, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28167630

RESUMEN

Ischemia-reperfusion injury (IRI) is an important cause of acute kidney injury that can lead to end-stage renal failure. Although the ensuing inflammatory response can restore homeostasis, a consecutive maladaptive repair and persistent inflammation represent important risk factors for postischemic chronic kidney disease development. In this study, we investigated the role of mast cells in both the early and late phases of the inflammatory response in experimental models of acute and chronic renal IRI using our recently developed mouse model that allows conditional ablation of mast cells. Depletion of mast cells prior to IRI resulted in improved renal function due to diminished local inflammatory cytokine/chemokine levels and neutrophil recruitment to the kidneys after the acute injury phase (48 h post-IRI). Furthermore, although not completely protected, mast cell-depleted mice displayed less organ atrophy and fibrosis than did wild-type mice during the chronic phases (2 and 6 wk post-IRI) of disease development. Conversely, mast cell ablation after the acute phase of IRI had no impact on organ atrophy, tubular necrosis, or fibrosis. Thus, our results suggest a deleterious role of mast cells during the acute inflammatory phase of IRI promoting subsequent fibrosis development, but not during the chronic phase of the disease.


Asunto(s)
Lesión Renal Aguda/inmunología , Riñón/inmunología , Mastocitos/inmunología , Daño por Reperfusión/inmunología , Animales , Degranulación de la Célula , Enfermedad Crónica , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Infiltración Neutrófila , Receptores de IgE/genética
11.
J Exp Med ; 213(7): 1353-74, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27353089

RESUMEN

Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit-independent MC-deficient (Cpa3(Cre/+)) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca(2+) desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force-Ca(2+) interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Mastocitos/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miofibrillas/metabolismo , Animales , Carboxipeptidasas A/genética , Carboxipeptidasas A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Ratones Noqueados , Contracción Miocárdica/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miofibrillas/patología , Proteolisis , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo
13.
Mol Immunol ; 63(1): 86-93, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24793464

RESUMEN

Mast cells are hematopoietic cells involved in inflammation and immunity and have been recognized also as important effector cells in kidney inflammation. In humans, only a few mast cells reside in kidneys constitutively but in progressive renal diseases their numbers increase substantially representing an essential part of the interstitial infiltrate of inflammatory cells. Recent data obtained in experimental animal models have emphasized a complex role of these cells and the mediators they release as they have been shown both to promote, but also to protect from disease and fibrosis development. Sometimes conflicting results have been reported in similar models suggesting a very narrow window between these activities depending on the pathophysiological context. Interestingly in mice, mast cell or mast cell mediator specific actions became also apparent in the absence of significant mast cell kidney infiltration supporting systemic or regional actions via draining lymph nodes or kidney capsules. Many of their activities rely on the capacity of mast cells to release, in a timely controlled manner, a wide range of inflammatory mediators, which can promote anti-inflammatory actions and repair activities that contribute to healing, but in some circumstances or in case of inappropriate regulation may also promote kidney disease.


Asunto(s)
Riñón/inmunología , Riñón/patología , Mastocitos/inmunología , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/patología , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Fibrosis , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Humanos , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Ratones
14.
J Clin Invest ; 124(10): 4577-89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25180604

RESUMEN

Controlling the overwhelming inflammatory reaction associated with polymicrobial sepsis remains a prevalent clinical challenge with few treatment options. In septic peritonitis, blood neutrophils and monocytes are rapidly recruited into the peritoneal cavity to control infection, but the role of resident sentinel cells during the early phase of infection is less clear. In particular, the influence of mast cells on other tissue-resident cells remains poorly understood. Here, we developed a mouse model that allows both visualization and conditional ablation of mast cells and basophils to investigate the role of mast cells in severe septic peritonitis. Specific depletion of mast cells led to increased survival rates in mice with acute sepsis. Furthermore, we determined that mast cells impair the phagocytic action of resident macrophages, thereby allowing local and systemic bacterial proliferation. Mast cells did not influence local recruitment of neutrophils and monocytes or the release of inflammatory cytokines. Phagocytosis inhibition by mast cells involved their ability to release prestored IL-4 within 15 minutes after bacterial encounter, and treatment with an IL-4-neutralizing antibody prevented this inhibitory effect and improved survival of septic mice. Our study uncovers a local crosstalk between mast cells and macrophages during the early phase of sepsis development that aggravates the outcome of severe bacterial infection.


Asunto(s)
Inflamación/inmunología , Macrófagos Peritoneales/citología , Mastocitos/citología , Fagocitosis , Sepsis/inmunología , Animales , Infecciones Bacterianas , Células de la Médula Ósea/citología , Proliferación Celular , Separación Celular , Citometría de Flujo , Interleucina-4/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Peritonitis/patología
15.
J Immunol ; 189(7): 3689-99, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22933633

RESUMEN

A favorable outcome following acute bacterial infection depends on the ability of phagocytic cells to be recruited and properly activated within injured tissues. Calcium (Ca(2+)) is a ubiquitous second messenger implicated in the functions of many cells, but the mechanisms involved in the regulation of Ca(2+) mobilization in hematopoietic cells are largely unknown. The monovalent cation channel transient receptor potential melastatin (TRPM) 4 is involved in the control of Ca(2+) signaling in some hematopoietic cell types, but the role of this channel in phagocytes and its relevance in the control of inflammation remain unexplored. In this study, we report that the ablation of the Trpm4 gene dramatically increased mouse mortality in a model of sepsis induced by cecal ligation and puncture. The lack of the TRPM4 channel affected macrophage population within bacteria-infected peritoneal cavities and increased the systemic level of Ly6C(+) monocytes and proinflammatory cytokine production. Impaired Ca(2+) mobilization in Trpm4(-/-) macrophages downregulated the AKT signaling pathway and the subsequent phagocytic activity, resulting in bacterial overgrowth and translocation to the bloodstream. In contrast, no alteration in the distribution, function, or Ca(2+) mobilization of Trpm4(-/-) neutrophils was observed, indicating that the mechanism controlling Ca(2+) signaling differs among phagocytes. Our results thus show that the tight control of Ca(2+) influx by the TRPM4 channel is critical for the proper functioning of monocytes/macrophages and the efficiency of the subsequent response to infection.


Asunto(s)
Macrófagos/inmunología , Macrófagos/patología , Monocitos/inmunología , Monocitos/patología , Neutrófilos , Sepsis/inmunología , Canales Catiónicos TRPM/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Humanos , Macrófagos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Peritonitis/inmunología , Peritonitis/metabolismo , Peritonitis/patología , Sepsis/metabolismo , Sepsis/patología , Canales Catiónicos TRPM/biosíntesis , Canales Catiónicos TRPM/deficiencia
16.
Front Immunol ; 2: 37, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22566827

RESUMEN

Mast cells are localized in tissues. Intense research on these cells over the years has demonstrated their role as effector cells in the maintenance of tissue integrity following injury produced by infectious agents, toxins, metabolic states, etc. After stimulation they release a sophisticated array of inflammatory mediators, cytokines, and growth factors to orchestrate an inflammatory response. These mediators can directly initiate tissue responses on resident cells, but they have also been shown to regulate other infiltrating immune cell functions. Research in recent years has revealed that the outcome of mast cell actions is not always detrimental for the host but can also limit disease development. In addition, mast cell functions highly depend on the physiological context in the organism. Depending on the genetic background, strength of the injurious event, the particular microenvironment, mast cells direct responses ranging from pro- to anti-inflammatory. It appears that they have evolved as cellular sensors to discern their environment in order to initiate an appropriate physiological response either aimed to favor inflammation for repair or at the contrary limit the inflammatory process to prevent further damage. Like every sophisticated machinery, its dysregulation leads to pathology. Given the broad distribution of mast cells in tissues this also explains their implication in many inflammatory diseases.

17.
Am J Pathol ; 168(2): 453-65, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16436660

RESUMEN

Originally implicated in axon guidance, semaphorins represent a large family of molecules that are now known to be expressed in the immune system. Among different semaphorins tested by reverse transcriptase-polymerase chain reaction in human immune cells, the expression of class 6 transmembrane semaphorin SEMA6A was restricted to dendritic cells (DCs). Using in-house generated monoclonal antibodies, SEMA6A expression appeared further restricted to Langerhans cells (LCs). In vivo, SEMA6A mRNA was expressed in freshly isolated skin LCs but SEMA6A protein was not detectable on normal skin and tonsillar epithelium. Of interest, SEMA6A protein was strongly expressed on skin and bone LCs and on LCs in draining lymph nodes from patients with LC histiocytosis or dermatopathic lymphadenitis, respectively, representing two inflammatory conditions in which LCs display an immature DC-LAMP(low), CD83(low), and CCR7+ phenotype. SEMA6A expression was low in resting LCs generated in vitro and was enhanced by interferon (IFN)-gamma but not by interleukin-4, interleukin-10, IFN-alpha/beta, or lipopolysaccharide. Most IFN-gamma-induced SEMA6A-positive cells remained immature with low CD83 and DC-LAMP/CD208 expression, but they expressed CCR7 and responded to macrophage inflammatory protein-3beta (MIP-3beta/CCL19). The expression of SEMA6A, for which the ligand and function remain unknown, may therefore identify an alternative IFN-gamma-dependent activation status of LCs in vivo.


Asunto(s)
Histiocitosis/metabolismo , Interferón gamma/farmacología , Células de Langerhans/metabolismo , Linfadenitis/metabolismo , Semaforinas/metabolismo , Adulto , Animales , Anticuerpos Monoclonales , Antígenos CD , Huesos/inmunología , Huesos/metabolismo , Huesos/patología , Encéfalo/metabolismo , Movimiento Celular , Quimiocina CCL19 , Quimiocinas CC/farmacología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/patología , Histiocitosis/patología , Humanos , Inmunoglobulinas , Interferón-alfa/farmacología , Interferón beta/farmacología , Interleucina-10/farmacología , Interleucina-4/farmacología , Células de Langerhans/inmunología , Lipopolisacáridos/farmacología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Linfadenitis/patología , Proteínas Inflamatorias de Macrófagos , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos BALB C , Tonsila Palatina/inmunología , Tonsila Palatina/metabolismo , Tonsila Palatina/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR7 , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Semaforinas/genética , Semaforinas/inmunología , Piel/inmunología , Piel/metabolismo , Piel/patología , Antígeno CD83
18.
J Exp Med ; 201(9): 1435-46, 2005 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-15851485

RESUMEN

Dendritic cells (DC) produce interleukin-12 (IL-12) in response to Toll-like receptor (TLR) activation. Two major TLR signaling pathways participate in the response to pathogens: the nuclear factor-kappaB (NF-kappaB)-dependent pathway leading to inflammatory cytokine secretion including IL-12 and the interferon (IFN)-dependent pathway inducing type I IFN and IFN-regulated genes. Here we show that the two pathways cooperate and are likely both necessary for inducing an optimal response to pathogens. R-848/Resiquimod (TLR7 ligand in the mouse and TLR7/8 ligand in human) synergized with poly(I:C) (TLR3 ligand) or lipopolysaccharide (LPS; TLR4 ligand) in inducing high levels of bioactive IL-12p70 secretion and IFN-beta mRNA accumulation by mouse bone marrow-derived DC (BM-DC). Strikingly, IL-12p70 but not IL-12p40 secretion was strongly reduced in BM-DC from STAT1(-/-) and IFNAR(-/-) mice. STAT1 tyrosine-phosphorylation, IL-12p35, and IFN-beta mRNA accumulation were strongly inhibited in IFNAR(-/-) BM-DC activated with the TLR ligand combinations. Similar observation were obtained in human TLR8-expressing monocyte-derived DC (moDC) using neutralizing anti-IFNAR2 antibodies, although results also pointed to a possible involvement of IFN-lambda1 (also known as IL-29). This suggests that TLR engagement on DC induces endogenous IFNs that further synergize with the NF-kappaB pathway for optimal IL-12p70 secretion. Moreover, analysis of interferon regulatory factors (IRF) regulation in moDC suggests a role for IRF7/8 in mediating IRF3-independent type I IFN and possibly IL-12p35 synthesis in response to TLR7/8.


Asunto(s)
Células Dendríticas/metabolismo , Interferón Tipo I/metabolismo , Interleucina-12/metabolismo , Glicoproteínas de Membrana/metabolismo , FN-kappa B/inmunología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/inmunología , Animales , Células de la Médula Ósea/metabolismo , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Imidazoles/metabolismo , Factor 3 Regulador del Interferón , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Receptor de Interferón alfa y beta , Receptores de Interferón/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1 , Receptor Toll-Like 3 , Receptor Toll-Like 4 , Receptor Toll-Like 7 , Receptor Toll-Like 8 , Receptores Toll-Like , Transactivadores/metabolismo , Factores de Transcripción
19.
Blood ; 102(13): 4431-40, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12907451

RESUMEN

Eph receptor tyrosine kinases and their ligands, the ephrins, have been primarily described in the nervous system for their roles in axon guidance, development, and cell intermingling. Here we address whether Eph receptors may also regulate dendritic cell (DC) trafficking. Reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that DCs derived from CD34+ progenitors, but not from monocytes, expressed several receptors, in particular EphA2, EphA4, EphA7, EphB1, and EphB3 mRNA. EphB3 was specifically expressed by Langerhans cells, and EphA2 and EphA7 were expressed by both Langerhans- and interstitial-type DCs. EphA and EphB protein expression on DCs generated in vitro was confirmed by staining with ephrin-A3-Fc and ephrin-B3-Fc fusion proteins that bind to different Eph members, in particular EphA2 and EphB3. Immunostaining with anti-EphA2 antibodies demonstrated the expression of EphA2 by immature DCs and by skin Langerhans cells isolated ex vivo. Interestingly, ephrin expression was detected in epidermal keratinocytes and also in DCs. Adhesion of CD34+-derived DCs to fibronectin, but not to poly-l-lysine, was increased in the presence of ephrin-A3-Fc, a ligand of EphA2, through a beta1 integrin activation pathway. As such, EphA2/ephrin-A3 interactions may play a role in the localization and network of Langerhans cells in the epithelium and in the regulation of their trafficking.


Asunto(s)
Células Dendríticas/enzimología , Efrina-A2/fisiología , Fibronectinas/química , Receptores de la Familia Eph/fisiología , Antígenos CD34/análisis , Adhesión Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Movimiento Celular , Células Dendríticas/citología , Efrina-A2/biosíntesis , Efrina-A2/genética , Efrina-A4/biosíntesis , Efrina-A4/genética , Efrina-B1/biosíntesis , Efrina-B1/genética , Efrina-B3/biosíntesis , Efrina-B3/genética , Células Epidérmicas , Epidermis/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Integrina beta1/fisiología , Queratinocitos/enzimología , Células de Langerhans/citología , Células de Langerhans/enzimología , Polilisina/química , Proteínas Recombinantes de Fusión/inmunología , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...