Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Annu Rev Cell Dev Biol ; 38: 467-489, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35850150

RESUMEN

Successful immune responses depend on the spatiotemporal coordination of immune cell migration, interactions, and effector functions in lymphoid and parenchymal tissues. Real-time intravital microscopy has revolutionized our understanding of the dynamic behavior of many immune cell types in the living tissues of several species. Observing immune cells in their native environment has revealed many unanticipated facets of their biology, which were not expected from experiments outside a living organism. Here we highlight both classic and more recent examples of surprising discoveries that critically relied on the use of live in vivo imaging. In particular, we focus on five major cell types of the innate immune response (macrophages, microglia, neutrophils, dendritic cells, and mast cells), and how studying their dynamics in mouse tissues has helped us advance our current knowledge of immune cell-mediated tissue homeostasis, host defense, and inflammation.


Asunto(s)
Inmunidad Innata , Microscopía Intravital , Animales , Inflamación , Microscopía Intravital/métodos , Macrófagos , Ratones
2.
Nat Immunol ; 24(6): 915-924, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37081147

RESUMEN

Immune cell locomotion is associated with amoeboid migration, a flexible mode of movement, which depends on rapid cycles of actin polymerization and actomyosin contraction1. Many immune cells do not necessarily require integrins, the major family of adhesion receptors in mammals, to move productively through three-dimensional tissue spaces2,3. Instead, they can use alternative strategies to transmit their actin-driven forces to the substrate, explaining their migratory adaptation to changing external environments4-6. However, whether these generalized concepts apply to all immune cells is unclear. Here, we show that the movement of mast cells (immune cells with important roles during allergy and anaphylaxis) differs fundamentally from the widely applied paradigm of interstitial immune cell migration. We identify a crucial role for integrin-dependent adhesion in controlling mast cell movement and localization to anatomical niches rich in KIT ligand, the major mast cell growth and survival factor. Our findings show that substrate-dependent haptokinesis is an important mechanism for the tissue organization of resident immune cells.


Asunto(s)
Actinas , Integrinas , Animales , Integrinas/metabolismo , Actinas/metabolismo , Mastocitos/metabolismo , Movimiento Celular , Leucocitos/metabolismo , Adhesión Celular , Mamíferos/metabolismo
3.
J Immunol ; 190(8): 4215-25, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23479227

RESUMEN

During experimental sepsis, excessive generation of the anaphylatoxin C5a results in reduction of the C5a receptor (C5aR) on neutrophils. These events have been shown to result in impaired innate immunity. However, the regulation and fate of C5aR on neutrophils during sepsis are largely unknown. In contrast to 30 healthy volunteers, 60 patients in septic shock presented evidence of complement activation with significantly increased serum levels of C3a, C5a, and C5b-9. In the septic shock group, the corresponding decrease in complement hemolytic activity distinguished survivors from nonsurvivors. Neutrophils from patients in septic shock exhibited decreased C5aR expression, which inversely correlated with serum concentrations of C-reactive protein (CRP) and clinical outcome. In vitro exposure of normal neutrophils to native pentameric CRP led to a dose- and time-dependent loss of C5aR expression on neutrophils, whereas the monomeric form of CRP, as well as various other inflammatory mediators, failed to significantly alter C5aR levels on neutrophils. A circulating form of C5aR (cC5aR) was detected in serum by immunoblotting and a flow-based capture assay, suggestive of an intact C5aR molecule. Levels of cC5aR were significantly enhanced during septic shock, with serum levels directly correlating with lethality. The data suggest that septic shock in humans is associated with extensive complement activation, CRP-dependent loss of C5aR on neutrophils, and appearance of cC5aR in serum, which correlated with a poor outcome. Therefore, cC5aR may represent a new sepsis marker to be considered in tailoring individualized immune-modulating therapy.


Asunto(s)
Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Complemento/sangre , Choque Séptico/sangre , Choque Séptico/inmunología , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptor de Anafilatoxina C5a , Receptores de Complemento/antagonistas & inhibidores , Choque Séptico/mortalidad , Sobrevida
4.
EMBO Rep ; 13(11): 1012-20, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-22964757

RESUMEN

Adipose tissue is the largest compartment in the mammalian body for storing energy as fat, providing an important reservoir of fuel for maintaining whole body energy homeostasis. Herein, we identify the transcriptional cofactor hairless (HR) to be required for white adipogenesis. Moreover, forced expression of HR in non-adipogenic precursor cells induces adipogenic gene expression and enhances adipocyte formation under permissive conditions. HR exerts its proadipogenic effects by regulating the expression of PPARγ, one of the central adipogenic transcription factors. In conclusion, our data provide a new mechanism required for white adipogenesis.


Asunto(s)
Adipocitos Blancos/citología , Adipogénesis/genética , Regulación del Desarrollo de la Expresión Génica , PPAR gamma/metabolismo , Factores de Transcripción/metabolismo , Células 3T3 , Adipocitos Blancos/metabolismo , Animales , Diferenciación Celular , Ratones , Ratones Noqueados , Mutación , PPAR gamma/genética , Factores de Transcripción/genética , Transcripción Genética
5.
J Immunol ; 187(8): 4374-83, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21930971

RESUMEN

Age-related macular degeneration is a leading form of blindness in Western countries and is associated with a common SNP (rs 1061170/Y402H) in the Factor H gene, which encodes the two complement inhibitors Factor H and FHL1. However, the functional consequences of this Tyr(402) His exchange in domain 7 are not precisely defined. In this study, we show that the Tyr(402) His sequence variation affects Factor H surface recruitment by monomeric C-reactive protein (mCRP) to specific patches on the surface of necrotic retinal pigment epithelial cells. Enhanced attachment of the protective Tyr(402) variants of both Factor H and FHL1 by mCRP results in more efficient complement control and further provides an anti-inflammatory environment. In addition, we demonstrate that mCRP is generated on the surface of necrotic retinal pigment epithelial cells and that this newly formed mCRP colocalizes with the cell damage marker annexin V. Bound to the cell surface, Factor H-mCRP complexes allow complement inactivation and reduce the release of the proinflammatory cytokine TNF-α. This mCRP-mediated complement inhibitory and anti-inflammatory activity at necrotic membrane lesions is affected by residue 402 of Factor H and defines a new role for mCRP, for Factor H, and also for the mCRP-Factor H complex. The increased protective capacity of the Tyr(402) Factor H variant allows better and more efficient clearance and removal of cellular debris and reduces inflammation and pathology.


Asunto(s)
Activación de Complemento/genética , Factor H de Complemento/genética , Degeneración Macular/genética , Degeneración Macular/patología , Proteína C-Reactiva/metabolismo , Separación Celular , Factor H de Complemento/química , Factor H de Complemento/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Degeneración Macular/metabolismo , Microscopía Confocal , Necrosis/genética , Polimorfismo de Nucleótido Simple , Unión Proteica , Estructura Cuaternaria de Proteína , Factores de Riesgo
6.
FASEB J ; 25(12): 4198-210, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21856781

RESUMEN

The acute-phase protein C-reactive protein (CRP) recruits C1q to the surface of damaged cells and thereby initiates complement activation. However, CRP also recruits complement inhibitors, such as C4b-binding protein (C4bp) and factor H, which both block complement progression at the level of C3 and inhibits inflammation. To define how CRP modulates the classic complement pathway, we studied the interaction of CRP with the classic pathway inhibitor C4bp. Monomeric CRP (mCRP), but not pentameric CRP (pCRP), binds C4bp and enhances degradation of C4b and C3b. Both C1q, the initiator, and C4bp, the inhibitor of the classic pathway, compete for mCRP binding, and this competition adjusts the local balance of activation and inhibition. After attachment of pCRP to the surface of necrotic rat myocytes, generation of mCRP was demonstrated over a period of 18 h. Similarly, a biological role for mCRP, C1q, and C4bp in the disease setting of acute myocardial infarction was revealed. In this inflamed tissue, mCRP, pCRP, C4bp, C1q, and C4d were detected in acetone-fixed and in unfixed tissue. Protein levels were enhanced 6 h to 5 d after infarction. Thus, mCRP bound to damaged cardiomyocytes recruits C1q to activate and also C4bp to control the classic complement pathway.


Asunto(s)
Proteína C-Reactiva/metabolismo , Vía Clásica del Complemento/fisiología , Animales , Proteína C-Reactiva/química , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Células Cultivadas , Complemento C1q/metabolismo , Proteína de Unión al Complemento C4b , Antígenos de Histocompatibilidad/metabolismo , Humanos , Células Jurkat , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Necrosis , Unión Proteica , Conformación Proteica , Estructura Cuaternaria de Proteína , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie
7.
Front Cell Dev Biol ; 10: 871789, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35478973

RESUMEN

Neutrophils are key cells of our innate immune response with essential roles for eliminating bacteria and fungi from tissues. They are also the prototype of an amoeboid migrating leukocyte. As one of the first blood-recruited immune cell types during inflammation and infection, these cells can invade almost any tissue compartment. Once in the tissue, neutrophils undergo rapid shape changes and migrate at speeds higher than most other immune cells. They move in a substrate-independent manner in interstitial spaces and do not follow predetermined tissue paths. Instead, neutrophil navigation is largely shaped by the chemokine and chemoattractant milieu around them. This highlights the decisive role of attractant-sensing G-protein coupled receptors (GPCRs) and downstream molecular pathways for controlling amoeboid neutrophil movement in tissues. A diverse repertoire of cell-surface expressed GPCRs makes neutrophils the perfect sentinel cell type to sense and detect danger-associated signals released from wounds, inflamed interstitium, dying cells, complement factors or directly from tissue-invading microbes. Moreover, neutrophils release attractants themselves, which allows communication and coordination between individual cells of a neutrophil population. GPCR-mediated positive feedback mechanisms were shown to underlie neutrophil swarming, a population response that amplifies the recruitment of amoeboid migrating neutrophils to sites of tissue injury and infection. Here we discuss recent findings and current concepts that counteract excessive neutrophil accumulation and swarm formation. In particular, we will focus on negative feedback control mechanisms that terminate neutrophil swarming to maintain the delicate balance between tissue surveillance, host protection and tissue destruction.

8.
Nat Commun ; 13(1): 1758, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35365619

RESUMEN

Fluorescence techniques dominate the field of live-cell microscopy, but bleaching and motion blur from too long integration times limit dynamic investigations of small objects. High contrast, label-free life-cell imaging of thousands of acquisitions at 160 nm resolution and 100 Hz is possible by Rotating Coherent Scattering (ROCS) microscopy, where intensity speckle patterns from all azimuthal illumination directions are added up within 10 ms. In combination with fluorescence, we demonstrate the performance of improved Total Internal Reflection (TIR)-ROCS with variable illumination including timescale decomposition and activity mapping at five different examples: millisecond reorganization of macrophage actin cortex structures, fast degranulation and pore opening in mast cells, nanotube dynamics between cardiomyocytes and fibroblasts, thermal noise driven binding behavior of virus-sized particles at cells, and, bacterial lectin dynamics at the cortex of lung cells. Using analysis methods we present here, we decipher how motion blur hides cellular structures and how slow structure motions cover decisive fast motions.


Asunto(s)
Actinas , Iluminación , Fibroblastos , Microscopía Fluorescente/métodos
9.
Nat Metab ; 4(7): 856-866, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35864246

RESUMEN

Successful elimination of bacteria in phagocytes occurs in the phago-lysosomal system, but also depends on mitochondrial pathways. Yet, how these two organelle systems communicate is largely unknown. Here we identify the lysosomal biogenesis factor transcription factor EB (TFEB) as regulator for phago-lysosome-mitochondria crosstalk in macrophages. By combining cellular imaging and metabolic profiling, we find that TFEB activation, in response to bacterial stimuli, promotes the transcription of aconitate decarboxylase (Acod1, Irg1) and synthesis of its product itaconate, a mitochondrial metabolite with antimicrobial activity. Activation of the TFEB-Irg1-itaconate signalling axis reduces the survival of the intravacuolar pathogen Salmonella enterica serovar Typhimurium. TFEB-driven itaconate is subsequently transferred via the Irg1-Rab32-BLOC3 system into the Salmonella-containing vacuole, thereby exposing the pathogen to elevated itaconate levels. By activating itaconate production, TFEB selectively restricts proliferating Salmonella, a bacterial subpopulation that normally escapes macrophage control, which contrasts TFEB's role in autophagy-mediated pathogen degradation. Together, our data define a TFEB-driven metabolic pathway between phago-lysosomes and mitochondria that restrains Salmonella Typhimurium burden in macrophages in vitro and in vivo.


Asunto(s)
Lisosomas , Succinatos , Autofagia/fisiología , Lisosomas/metabolismo , Macrófagos/metabolismo , Succinatos/metabolismo , Succinatos/farmacología
10.
Blood ; 114(12): 2439-47, 2009 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-19528535

RESUMEN

Homozygous deletion of a 84-kb genomic fragment in human chromosome 1 that encompasses the CFHR1 and CFHR3 genes represents a risk factor for hemolytic uremic syndrome (HUS) but has a protective effect in age-related macular degeneration (AMD). Here we identify CFHR1 as a novel inhibitor of the complement pathway that blocks C5 convertase activity and interferes with C5b surface deposition and MAC formation. This activity is distinct from complement factor H, and apparently factor H and CFHR1 control complement activation in a sequential manner. As both proteins bind to the same or similar sites at the cellular surfaces, the gain of CFHR1 activity presumably is at the expense of CFH-mediated function (inhibition of the C3 convertase). In HUS, the absence of CFHR1 may result in reduced inhibition of terminal complex formation and in reduced protection of endothelial cells upon complement attack. These findings provide new insights into complement regulation on the cell surface and biosurfaces and likely define the role of CFHR1 in human diseases.


Asunto(s)
Convertasas de Complemento C3-C5/antagonistas & inhibidores , Proteínas Inactivadoras del Complemento C3b/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Animales , Células Cultivadas , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Complemento C3d/metabolismo , Factor H de Complemento/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Microscopía Confocal , Retina/citología , Retina/metabolismo , Ovinos , Venas Umbilicales/citología , Venas Umbilicales/metabolismo
11.
Curr Opin Cell Biol ; 72: 156-162, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34500367

RESUMEN

Several immune cell types (neutrophils, eosinophils, T cells, and innate-like lymphocytes) display coordinated migration patterns when a population, formed of individually responding cells, moves through inflamed or infected tissues. "Swarming" refers to the process in which a population of migrating leukocytes switches from random motility to highly directed chemotaxis to form local cell clusters. Positive feedback amplification underlies this behavior and results from intercellular communication in the immune cell population. We here highlight recent findings on neutrophil swarming from mouse models, zebrafish larvae, and in vitro platforms for human cells, which together advanced our understanding of the principles and molecular mechanisms that shape immune cell swarming.


Asunto(s)
Neutrófilos , Pez Cebra , Animales , Quimiotaxis , Retroalimentación , Ratones
12.
Science ; 372(6548)2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34140358

RESUMEN

Neutrophils communicate with each other to form swarms in infected organs. Coordination of this population response is critical for the elimination of bacteria and fungi. Using transgenic mice, we found that neutrophils have evolved an intrinsic mechanism to self-limit swarming and avoid uncontrolled aggregation during inflammation. G protein-coupled receptor (GPCR) desensitization acts as a negative feedback control to stop migration of neutrophils when they sense high concentrations of self-secreted attractants that initially amplify swarming. Interference with this process allows neutrophils to scan larger tissue areas for microbes. Unexpectedly, this does not benefit bacterial clearance as containment of proliferating bacteria by neutrophil clusters becomes impeded. Our data reveal how autosignaling stops self-organized swarming behavior and how the finely tuned balance of neutrophil chemotaxis and arrest counteracts bacterial escape.


Asunto(s)
Quimiotaxis de Leucocito , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Ganglios Linfáticos/microbiología , Neutrófilos/fisiología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/crecimiento & desarrollo , Animales , Agregación Celular , Quimiocina CXCL2 , Eosinófilos/fisiología , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Inflamación , Leucotrieno B4/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neutrófilos/inmunología , Infecciones por Pseudomonas/microbiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Piel/inmunología , Piel/lesiones , Piel/patología
13.
Mol Immunol ; 46(3): 335-44, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19084272

RESUMEN

Human complement factor H-related protein 4 (CFHR4) is a plasma glycoprotein which appears in two isoforms. CFHR4 is a member of the factor H protein family, and shares structural similarity and sequence homology with the other CFHR proteins and with the complement regulator factor H. Given the structural and sequence similarity, we hypothesized that similar to factor H, CFHR4 binds to C-reactive protein (CRP). We have recombinantly expressed the two CFHR4 isoforms and analyzed their binding to both native and denatured, monomeric CRP. Here, we show that both CFHR4 isoforms bind in the presence of calcium to native pentameric CRP, but not to modified CRP. This is in contrast to factor H, which binds to modified CRP independent of calcium. Comparison of the two CFHR4 isoforms and a recombinant CFHR4 fragment for CRP binding indicates that the first domain of CFHR4 is relevant for this interaction. Interaction of the native proteins was demonstrated by co-precipitation of CFHR4 and CRP from serum of sepsis patients with elevated CRP levels. CFHR4 bound to necrotic cells and was localized in necrotic tumor tissue as demonstrated by immunohistological analyses. In addition, CFHR4 facilitated binding of native CRP to the surface of necrotic cells. Altogether these data identify CFHR4 as a novel ligand for native CRP, and suggest a role for CFHR4 in opsonization of necrotic cells.


Asunto(s)
Apolipoproteínas/metabolismo , Proteína C-Reactiva/metabolismo , Necrosis/metabolismo , Apolipoproteínas/química , Apolipoproteínas/aislamiento & purificación , Tampones (Química) , Calcio/farmacología , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Neoplasias del Colon/patología , Humanos , Inmunoprecipitación , Concentración Osmolar , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/química , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
14.
J Exp Med ; 214(9): 2671-2693, 2017 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-28716882

RESUMEN

The inflammasomes are multiprotein complexes sensing tissue damage and infectious agents to initiate innate immune responses. Different inflammasomes containing distinct sensor molecules exist. The NLRP3 inflammasome is unique as it detects a variety of danger signals. It has been reported that NLRP3 is recruited to mitochondria-associated endoplasmic reticulum membranes (MAMs) and is activated by MAM-derived effectors. Here, we show that in response to inflammasome activators, MAMs localize adjacent to Golgi membranes. Diacylglycerol (DAG) at the Golgi rapidly increases, recruiting protein kinase D (PKD), a key effector of DAG. Upon PKD inactivation, self-oligomerized NLRP3 is retained at MAMs adjacent to Golgi, blocking assembly of the active inflammasome. Importantly, phosphorylation of NLRP3 by PKD at the Golgi is sufficient to release NLRP3 from MAMs, resulting in assembly of the active inflammasome. Moreover, PKD inhibition prevents inflammasome autoactivation in peripheral blood mononuclear cells from patients carrying NLRP3 mutations. Hence, Golgi-mediated PKD signaling is required and sufficient for NLRP3 inflammasome activation.


Asunto(s)
Aparato de Golgi/fisiología , Inflamasomas/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Proteína Quinasa C/fisiología , Animales , Diglicéridos/metabolismo , Retículo Endoplásmico/fisiología , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación
15.
Cell Death Dis ; 7(10): e2411, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27735945

RESUMEN

Adaptation to changes in nutrient availability is crucial for cells and organisms. Posttranslational modifications of signaling proteins are very dynamic and are therefore key to promptly respond to nutrient deprivation or overload. Herein we screened for ubiquitylation of proteins in the livers of fasted and refed mice using a comprehensive systemic proteomic approach. Among 1641 identified proteins, 117 were differentially ubiquitylated upon fasting or refeeding. Endoplasmic reticulum (ER) and secretory proteins were enriched in the livers of refed mice in part owing to an ER-stress-mediated response engaging retro-translocation and ubiquitylation of proteins from the ER. Complement C3, an innate immune factor, emerged as the most prominent ER-related hit of our screen. Accordingly, we found that secretion of C3 from the liver and primary hepatocytes as well as its dynamic trafficking are nutrient dependent. Finally, obese mice with a chronic nutrient overload show constitutive trafficking of C3 in the livers despite acute changes in nutrition, which goes in line with increased C3 levels and low-grade inflammation reported for obese patients. Our study thus suggests that nutrient sensing in the liver is coupled to release of C3 and potentially its metabolic and inflammatory functions.


Asunto(s)
Complemento C3/metabolismo , Hígado/metabolismo , Proteoma/metabolismo , Ubiquitinas/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Conducta Alimentaria , Células HEK293 , Humanos , Espacio Intracelular/metabolismo , Ratones Endogámicos C57BL , Transporte de Proteínas , Vesículas Secretoras/metabolismo , Estrés Fisiológico , Ubiquitinación
16.
Science ; 347(6224): 878-82, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25700520

RESUMEN

Pancreatic ß cells lower insulin release in response to nutrient depletion. The question of whether starved ß cells induce macroautophagy, a predominant mechanism maintaining energy homeostasis, remains poorly explored. We found that, in contrast to many mammalian cells, macroautophagy in pancreatic ß cells was suppressed upon starvation. Instead, starved ß cells induced lysosomal degradation of nascent secretory insulin granules, which was controlled by protein kinase D (PKD), a key player in secretory granule biogenesis. Starvation-induced nascent granule degradation triggered lysosomal recruitment and activation of mechanistic target of rapamycin that suppressed macroautophagy. Switching from macroautophagy to insulin granule degradation was important to keep insulin secretion low upon fasting. Thus, ß cells use a PKD-dependent mechanism to adapt to nutrient availability and couple autophagy flux to secretory function.


Asunto(s)
Autofagia , Células Secretoras de Insulina/fisiología , Insulina/metabolismo , Vesículas Secretoras/fisiología , Animales , Células Cultivadas , Ayuno , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Ratones , Ratones Mutantes , Ratones Transgénicos , Proteína Quinasa 13 Activada por Mitógenos/genética , Proteína Quinasa C/fisiología , Vesículas Secretoras/metabolismo
18.
PLoS One ; 7(2): e31223, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22319619

RESUMEN

Invasive bronchopulmonary aspergillosis (IBPA) is a life-threatening disease in immunocompromised patients. Although Aspergillus terreus is frequently found in the environment, A. fumigatus is by far the main cause of IBPA. However, once A. terreus establishes infection in the host, disease is as fatal as A. fumigatus infections. Thus, we hypothesized that the initial steps of disease establishment might be fundamentally different between these two species. Since alveolar macrophages represent one of the first phagocytes facing inhaled conidia, we compared the interaction of A. terreus and A. fumigatus conidia with alveolar macrophages. A. terreus conidia were phagocytosed more rapidly than A. fumigatus conidia, possibly due to higher exposure of ß-1,3-glucan and galactomannan on the surface. In agreement, blocking of dectin-1 and mannose receptors significantly reduced phagocytosis of A. terreus, but had only a moderate effect on phagocytosis of A. fumigatus. Once phagocytosed, and in contrast to A. fumigatus, A. terreus did not inhibit acidification of phagolysosomes, but remained viable without signs of germination both in vitro and in immunocompetent mice. The inability of A. terreus to germinate and pierce macrophages resulted in significantly lower cytotoxicity compared to A. fumigatus. Blocking phagolysosome acidification by the v-ATPase inhibitor bafilomycin increased A. terreus germination rates and cytotoxicity. Recombinant expression of the A. nidulans wA naphthopyrone synthase, a homologue of A. fumigatus PksP, inhibited phagolysosome acidification and resulted in increased germination, macrophage damage and virulence in corticosteroid-treated mice. In summary, we show that A. terreus and A. fumigatus have evolved significantly different strategies to survive the attack of host immune cells. While A. fumigatus prevents phagocytosis and phagolysosome acidification and escapes from macrophages by germination, A. terreus is rapidly phagocytosed, but conidia show long-term persistence in macrophages even in immunocompetent hosts.


Asunto(s)
Aspergillus fumigatus/patogenicidad , Aspergillus/patogenicidad , Macrófagos Alveolares/microbiología , Aspergilosis Pulmonar/microbiología , Animales , Ratones , Fagocitosis , Esporas Fúngicas
19.
Int J Oncol ; 39(1): 161-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21503575

RESUMEN

Human complement factor H (CFH), a central complement control protein, is a member of the regulators of complement activation family. Recent studies suggested that CFH may play a key role in the resistance of complement-mediated lysis in various cancer cells. In this study, we investigated the role of CFH in human lung cancer. Expression of CFH was analyzed in lung cancer cell lines by RT-PCR, Western blotting and immunofluorescence. In primary lung tumors, the protein expression of CFH was evaluated by immunohistochemistry (IHC) on tissue microarray (TMA). Binding of CFH to lung cancer cells was detected by flow cytometry. mRNA expression of CFH was detected in 6 out of 10 non-small cell lung cancer (NSCLC) cell lines, but in none of the small cell lung cancer (SCLC) cell lines. In line with Western blotting, immunofluorescence analysis demonstrated CFH protein expression in 3 NSCLC cell lines, and the immunoreaction was mainly associated with cell cytoplasm and membrane. In primary lung tumors, 54 out of 101 samples exhibited high expression of CFH and high expression was significantly correlated with lung adenocarcinoma (p=0.009). Also, in adenocarcinoma of the lung, Kaplan-Meier survival analysis showed a tendency that CFH-positive tumors had worse prognosis in comparison to CFH-negative tumors (p=0.082). Additionally, shorter survival time of patients with adenocarcinoma (<20 months) was associated with higher staining of CFH (p=0.033). Our data showed that non-small cell lung cancer cells expressed and secreted CFH. CFH might be a novel diagnostic marker for human lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/diagnóstico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Neoplasias Pulmonares/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/patología , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Estadificación de Neoplasias , Unión Proteica , Análisis de Supervivencia
20.
Mol Immunol ; 47(6): 1347-55, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20042240

RESUMEN

C-reactive protein (CRP) is a pattern recognition molecule that binds several microbial and host ligands. Ligand-bound CRP activates the complement system via the classical pathway. Previously, we identified human complement factor H-related protein 4 (CFHR4), a member of the factor H protein family, as a CRP binding protein. Here, we investigated the molecular basis and the functional relevance of the interaction of CFHR4 with native CRP. Using recombinantly expressed CFHR4 fragments, the CRP binding site was localized to the first short consensus repeat (SCR) domain of CFHR4. Peptide arrays identified residues 35-41 of CFHR4 to be involved in CRP binding. Substitutions of the positively charged amino acids of this motif resulted in strongly reduced CRP binding. Sequence comparisons revealed that such a motif is not present in the related SCR6 domain of factor H, or in the homologous domains of the four other CFHR proteins. Homology modelling based on SCR6 of factor H showed that the CRP binding site is surface exposed on SCR1 of CFHR4. CFHR4-bound CRP was able to activate complement, determined by C3 fragment deposition. Recombinant CFHR4 proteins with mutations in the identified binding site showed reduced CRP binding, which in turn resulted in reduced complement activation. In summary, these data reveal the molecular basis of the specific interaction of CFHR4 with native CRP and suggest a role for CFHR4 in enhancing opsonization via CRP binding.


Asunto(s)
Apolipoproteínas/química , Apolipoproteínas/inmunología , Proteína C-Reactiva/metabolismo , Activación de Complemento/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Aminoácidos/farmacología , Sitios de Unión , Activación de Complemento/efectos de los fármacos , Complemento C3/inmunología , Secuencia de Consenso , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Análisis por Matrices de Proteínas , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Secuencias Repetitivas de Aminoácido , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA