Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Immunity ; 54(9): 1989-2004.e9, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34363750

RESUMEN

The migration of neutrophils from the blood circulation to sites of infection or injury is a key immune response and requires the breaching of endothelial cells (ECs) that line the inner aspect of blood vessels. Unregulated neutrophil transendothelial cell migration (TEM) is pathogenic, but the molecular basis of its physiological termination remains unknown. Here, we demonstrated that ECs of venules in inflamed tissues exhibited a robust autophagic response that was aligned temporally with the peak of neutrophil trafficking and was strictly localized to EC contacts. Genetic ablation of EC autophagy led to excessive neutrophil TEM and uncontrolled leukocyte migration in murine inflammatory models, while pharmacological induction of autophagy suppressed neutrophil infiltration into tissues. Mechanistically, autophagy regulated the remodeling of EC junctions and expression of key EC adhesion molecules, facilitating their intracellular trafficking and degradation. Collectively, we have identified autophagy as a modulator of EC leukocyte trafficking machinery aimed at terminating physiological inflammation.


Asunto(s)
Autofagia/fisiología , Células Endoteliales/fisiología , Infiltración Neutrófila/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Quimiotaxis de Leucocito/fisiología , Células Endoteliales/patología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Uniones Intercelulares/fisiología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología
2.
Immunity ; 54(7): 1494-1510.e7, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34033752

RESUMEN

Aging is associated with dysregulated immune functions. Here, we investigated the impact of age on neutrophil diapedesis. Using confocal intravital microscopy, we found that in aged mice, neutrophils adhered to vascular endothelium in inflamed tissues but exhibited a high frequency of reverse transendothelial migration (rTEM). This retrograde breaching of the endothelium by neutrophils was governed by enhanced production of the chemokine CXCL1 from mast cells that localized at endothelial cell (EC) junctions. Increased EC expression of the atypical chemokine receptor 1 (ACKR1) supported this pro-inflammatory milieu in aged venules. Accumulation of CXCL1 caused desensitization of the chemokine receptor CXCR2 on neutrophils and loss of neutrophil directional motility within EC junctions. Fluorescent tracking revealed that in aged mice, neutrophils undergoing rTEM re-entered the circulation and disseminated to the lungs where they caused vascular leakage. Thus, neutrophils stemming from a local inflammatory site contribute to remote organ damage, with implication to the dysregulated systemic inflammation associated with aging.


Asunto(s)
Envejecimiento/inmunología , Transporte Biológico/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Animales , Quimiocina CXCL1/inmunología , Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Femenino , Uniones Intercelulares/inmunología , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-8B/inmunología , Vénulas/inmunología
3.
Immunity ; 49(6): 1062-1076.e6, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30446388

RESUMEN

Neutrophils require directional cues to navigate through the complex structure of venular walls and into inflamed tissues. Here we applied confocal intravital microscopy to analyze neutrophil emigration in cytokine-stimulated mouse cremaster muscles. We identified differential and non-redundant roles for the chemokines CXCL1 and CXCL2, governed by their distinct cellular sources. CXCL1 was produced mainly by TNF-stimulated endothelial cells (ECs) and pericytes and supported luminal and sub-EC neutrophil crawling. Conversely, neutrophils were the main producers of CXCL2, and this chemokine was critical for correct breaching of endothelial junctions. This pro-migratory activity of CXCL2 depended on the atypical chemokine receptor 1 (ACKR1), which is enriched within endothelial junctions. Transmigrating neutrophils promoted a self-guided migration response through EC junctions, creating a junctional chemokine "depot" in the form of ACKR1-presented CXCL2 that enabled efficient unidirectional luminal-to-abluminal migration. Thus, CXCL1 and CXCL2 act in a sequential manner to guide neutrophils through venular walls as governed by their distinct cellular sources.


Asunto(s)
Quimiocina CXCL1 , Quimiocina CXCL2 , Sistema del Grupo Sanguíneo Duffy , Neutrófilos , Receptores de Superficie Celular , Migración Transendotelial y Transepitelial , Animales , Músculos Abdominales/efectos de los fármacos , Músculos Abdominales/inmunología , Músculos Abdominales/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Quimiocina CXCL2/metabolismo , Sistema del Grupo Sanguíneo Duffy/genética , Sistema del Grupo Sanguíneo Duffy/inmunología , Sistema del Grupo Sanguíneo Duffy/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/inmunología , Uniones Intercelulares/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/genética , Migración Transendotelial y Transepitelial/inmunología , Factor de Necrosis Tumoral alfa/farmacología
4.
EMBO Rep ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918502

RESUMEN

Cellular senescence is a hallmark of advanced age and a major instigator of numerous inflammatory pathologies. While endothelial cell (EC) senescence is aligned with defective vascular functionality, its impact on fundamental inflammatory responses in vivo at single-cell level remain unclear. To directly investigate the role of EC senescence on dynamics of neutrophil-venular wall interactions, we applied high resolution confocal intravital microscopy to inflamed tissues of an EC-specific progeroid mouse model, characterized by profound indicators of EC senescence. Progerin-expressing ECs supported prolonged neutrophil adhesion and crawling in a cell autonomous manner that additionally mediated neutrophil-dependent microvascular leakage. Transcriptomic and immunofluorescence analysis of inflamed tissues identified elevated levels of EC CXCL1 on progerin-expressing ECs and functional blockade of CXCL1 suppressed the dysregulated neutrophil responses elicited by senescent ECs. Similarly, cultured progerin-expressing human ECs exhibited a senescent phenotype, were pro-inflammatory and prompted increased neutrophil attachment and activation. Collectively, our findings support the concept that senescent ECs drive excessive inflammation and provide new insights into the mode, dynamics, and mechanisms of this response at single-cell level.

5.
Proc Natl Acad Sci U S A ; 120(17): e2211631120, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37071676

RESUMEN

Fibromyalgia is a debilitating widespread chronic pain syndrome that occurs in 2 to 4% of the population. The prevailing view that fibromyalgia results from central nervous system dysfunction has recently been challenged with data showing changes in peripheral nervous system activity. Using a mouse model of chronic widespread pain through hyperalgesic priming of muscle, we show that neutrophils invade sensory ganglia and confer mechanical hypersensitivity on recipient mice, while adoptive transfer of immunoglobulin, serum, lymphocytes, or monocytes has no effect on pain behavior. Neutrophil depletion abolishes the establishment of chronic widespread pain in mice. Neutrophils from patients with fibromyalgia also confer pain on mice. A link between neutrophil-derived mediators and peripheral nerve sensitization is already established. Our observations suggest approaches for targeting fibromyalgia pain via mechanisms that cause altered neutrophil activity and interactions with sensory neurons.


Asunto(s)
Dolor Crónico , Fibromialgia , Humanos , Neutrófilos , Hiperalgesia , Ganglios Sensoriales
6.
Nat Immunol ; 12(8): 761-9, 2011 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-21706006

RESUMEN

The migration of neutrophils into inflamed tissues is a fundamental component of innate immunity. A decisive step in this process is the polarized migration of blood neutrophils through endothelial cells (ECs) lining the venular lumen (transendothelial migration (TEM)) in a luminal-to-abluminal direction. By real-time confocal imaging, we found that neutrophils had disrupted polarized TEM ('hesitant' and 'reverse') in vivo. We noted these events in inflammation after ischemia-reperfusion injury, characterized by lower expression of junctional adhesion molecule C (JAM-C) at EC junctions, and they were enhanced by blockade or genetic deletion of JAM-C in ECs. Our results identify JAM-C as a key regulator of polarized neutrophil TEM in vivo and suggest that reverse TEM of neutrophils can contribute to the dissemination of systemic inflammation.


Asunto(s)
Moléculas de Adhesión Celular/inmunología , Endotelio Vascular/inmunología , Inmunoglobulinas/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Migración Transendotelial y Transepitelial/inmunología , Animales , Endotelio Vascular/citología , Endotelio Vascular/patología , Procesamiento de Imagen Asistido por Computador , Inflamación/patología , Ratones , Microscopía Confocal , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología
7.
FASEB J ; 36(1): e22065, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34847625

RESUMEN

Neutrophil trafficking is a key component of the inflammatory response. Here, we have investigated the role of the immunomodulatory lectin Galectin-9 (Gal-9) on neutrophil recruitment. Our data indicate that Gal-9 is upregulated in the inflamed vasculature of RA synovial biopsies and report the release of Gal-9 into the extracellular environment following endothelial cell activation. siRNA knockdown of endothelial Gal-9 resulted in reduced neutrophil adhesion and neutrophil recruitment was significantly reduced in Gal-9 knockout mice in a model of zymosan-induced peritonitis. We also provide evidence for Gal-9 binding sites on human neutrophils; Gal-9 binding induced neutrophil activation (increased expression of ß2 integrins and reduced expression of CD62L). Intra-vital microscopy confirmed a pro-recruitment role for Gal-9, with increased numbers of transmigrated neutrophils following Gal-9 administration. We studied the role of both soluble and immobilized Gal-9 on human neutrophil recruitment. Soluble Gal-9 significantly strengthened the interaction between neutrophils and the endothelium and inhibited neutrophil crawling on ICAM-1. When immobilized, Gal-9 functioned as an adhesion molecule and captured neutrophils from the flow. Neutrophils adherent to Gal-9 exhibited a spread/activated phenotype that was inhibited by CD18 and CD44 neutralizing antibodies, suggesting a role for these molecules in the pro-adhesive effects of Gal-9. Our data indicate that Gal-9 is expressed and released by the activated endothelium and functions both in soluble form and when immobilized as a neutrophil adhesion molecule. This study paves the way for further investigation of the role of Gal-9 in leukocyte recruitment in different inflammatory settings.


Asunto(s)
Antígenos CD18/metabolismo , Galectinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Receptores de Hialuranos/metabolismo , Neutrófilos/metabolismo , Migración Transendotelial y Transepitelial , Animales , Adhesión Celular , Humanos , Ratones
8.
J Pathol ; 247(5): 662-671, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30584795

RESUMEN

Recent advances have provided evidence for the involvement of neutrophils in both innate and adaptive immunity, robustly challenging the old dogma that neutrophils are short-lived prototypical innate immune cells solely involved in acute responses to microbes and exerting collateral tissue damage. There is now ample evidence showing that neutrophils can migrate into different compartments of the lymphoid system where they contribute to the orchestration of the activation and/or suppression of lymphocyte effector functions in homeostasis and during chronic inflammation, such as autoimmune disorders and cancer. In support of this notion, neutrophils can generate a wide range of cytokines and other mediators capable of regulating the survival, proliferation and functions of both T and B cells. In addition, neutrophils can directly engage with lymphocytes and promote antigen presentation. Furthermore, there is emerging evidence of the existence of distinct and diverse neutrophil phenotypes with immunomodulatory functions that characterise different pathological conditions, including chronic and autoimmune inflammatory conditions. The aim of this review is to discuss the mechanisms implicated in neutrophil trafficking into the lymphoid system and to provide an overview of the immuno-regulatory functions of neutrophils in health and disease in the context of adaptive immunity. Copyright © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Ganglios Linfáticos/fisiología , Neoplasias/inmunología , Neutrófilos/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Humanos , Fenotipo
9.
J Pathol ; 248(1): 88-102, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30632166

RESUMEN

Ischemia/reperfusion (I/R) injury is a severe inflammatory insult associated with numerous pathologies, such as myocardial infarction, stroke and acute kidney injury. I/R injury is characterized by a rapid influx of activated neutrophils secreting toxic free radical species and degrading enzymes that can irreversibly damage the tissue, thus impairing organ functions. Significant efforts have been invested in identifying therapeutic targets to suppress neutrophil recruitment and activation post-I/R injury. In this context, pharmacological targeting of neutrophil elastase (NE) has shown promising anti-inflammatory efficacy in a number of experimental and clinical settings of I/R injury and is considered a plausible clinical strategy for organ care. However, the mechanisms of action of NE, and hence its inhibitors, in this process are not fully understood. Here we conducted a comprehensive analysis of the impact of NE genetic deletion on neutrophil infiltration in four murine models of I/R injury as induced in the heart, kidneys, intestine and cremaster muscle. In all models, neutrophil migration into ischemic regions was significantly suppressed in NE-/- mice as compared with wild-type controls. Analysis of inflamed cremaster muscle and mesenteric microvessels by intravital and confocal microscopy revealed a selective entrapment of neutrophils within venular walls, most notably at the level of the venular basement membrane (BM) following NE deletion/pharmacological blockade. This effect was associated with the suppression of NE-mediated remodeling of the low matrix protein expressing regions within the venular BM used by transmigrating neutrophils as exit portals. Furthermore, whilst NE deficiency led to reduced neutrophil activation and vascular leakage, levels of monocytes and prohealing M2 macrophages were reduced in tissues of NE-/- mice subjected to I/R. Collectively our results identify a vital and non-redundant role for NE in supporting neutrophil breaching of the venular BM post-I/R injury but also suggest a protective role for NE in promoting tissue repair. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Elastasa de Leucocito/fisiología , Neutrófilos/fisiología , Daño por Reperfusión/enzimología , Migración Transendotelial y Transepitelial/fisiología , Remodelación Vascular/fisiología , Animales , Membrana Basal/enzimología , Membrana Basal/patología , Membrana Basal/fisiopatología , Modelos Animales de Enfermedad , Eliminación de Gen , Riñón/irrigación sanguínea , Riñón/patología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Infiltración Neutrófila/fisiología , Neutrófilos/enzimología , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Vénulas/enzimología , Vénulas/patología , Vénulas/fisiopatología
10.
Blood ; 127(7): 898-907, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26647392

RESUMEN

Intracellular adhesion molecule-1 (ICAM-1) is a transmembrane glycoprotein expressed on the cell surface of numerous cell types such as endothelial and epithelial cells, vascular smooth muscle cells, and certain leukocyte subsets. With respect to the latter, ICAM-1 has been detected on neutrophils in several clinical and experimental settings, but little is known about the regulation of expression or function of neutrophil ICAM-1. In this study, we report on the de novo induction of ICAM-1 on the cell surface of murine neutrophils by lipopolysaccharide (LPS), tumor necrosis factor, and zymosan particles in vitro. The induction of neutrophil ICAM-1 was associated with enhanced phagocytosis of zymosan particles and reactive oxygen species (ROS) generation. Conversely, neutrophils from ICAM-1-deficient mice were defective in these effector functions. Mechanistically, ICAM-1-mediated intracellular signaling appeared to support neutrophil ROS generation and phagocytosis. In vivo, LPS-induced inflammation in the mouse cremaster muscle and peritoneal cavity led to ICAM-1 expression on intravascular and locally transmigrated neutrophils. The use of chimeric mice deficient in ICAM-1 on myeloid cells demonstrated that neutrophil ICAM-1 was not required for local neutrophil transmigration, but supported optimal intravascular and extravascular phagocytosis of zymosan particles. Collectively, the present results shed light on regulation of expression and function of ICAM-1 on neutrophils and identify it as an additional regulator of neutrophil effector responses in host defense.


Asunto(s)
Endotoxemia/inducido químicamente , Endotoxemia/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/biosíntesis , Lipopolisacáridos/toxicidad , Neutrófilos/metabolismo , Animales , Modelos Animales de Enfermedad , Endotoxemia/genética , Endotoxemia/patología , Molécula 1 de Adhesión Intercelular/genética , Ratones , Ratones Noqueados , Neutrófilos/patología , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Especies Reactivas de Oxígeno/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/genética
11.
Mediators Inflamm ; 2015: 946509, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26568666

RESUMEN

Leukocyte extravasation is one of the essential and first steps during the initiation of inflammation. Therefore, a better understanding of the key molecules that regulate this process may help to develop novel therapeutics for treatment of inflammation-based diseases such as atherosclerosis or rheumatoid arthritis. The endothelial adhesion molecules ICAM-1 and VCAM-1 are known as the central mediators of leukocyte adhesion to and transmigration across the endothelium. Engagement of these molecules by their leukocyte integrin receptors initiates the activation of several signaling pathways within both leukocytes and endothelium. Several of such events have been described to occur during transendothelial migration of all leukocyte subsets, whereas other mechanisms are known only for a single leukocyte subset. Here, we summarize current knowledge on regulatory mechanisms of leukocyte extravasation from a leukocyte and endothelial point of view, respectively. Specifically, we will focus on highlighting common and unique mechanisms that specific leukocyte subsets exploit to succeed in crossing endothelial monolayers.


Asunto(s)
Leucocitos/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/fisiología , Humanos , Inflamación/etiología , Inflamación/patología , Inflamación/fisiopatología , Molécula 1 de Adhesión Intercelular/fisiología , Leucocitos/clasificación , Ratones , Modelos Biológicos , Molécula 1 de Adhesión Celular Vascular/fisiología
12.
J Exp Med ; 203(6): 1519-32, 2006 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-16754715

RESUMEN

The mechanism of leukocyte migration through venular walls in vivo is largely unknown. By using immunofluorescence staining and confocal microscopy, the present study demonstrates the existence of regions within the walls of unstimulated murine cremasteric venules where expression of key vascular basement membrane (BM) constituents, laminin 10, collagen IV, and nidogen-2 (but not perlecan) are considerably lower (<60%) than the average expression detected in the same vessel. These sites were closely associated with gaps between pericytes and were preferentially used by migrating neutrophils during their passage through cytokine-stimulated venules. Although neutrophil transmigration did not alter the number/unit area of extracellular matrix protein low expression sites, the size of these regions was enlarged and their protein content was reduced in interleukin-1beta-stimulated venules. These effects were entirely dependent on the presence of neutrophils and appeared to involve neutrophil-derived serine proteases. Furthermore, evidence was obtained indicating that transmigrating neutrophils carry laminins on their cell surface in vivo. Collectively, through identification of regions of low extracellular matrix protein localization that define the preferred route for transmigrating neutrophils, we have identified a plausible mechanism by which neutrophils penetrate the vascular BM without causing a gross disruption to its intricate structure.


Asunto(s)
Membrana Basal/fisiología , Proteínas de la Matriz Extracelular/genética , Neutrófilos/fisiología , Vénulas/fisiología , Animales , Movimiento Celular , Cartilla de ADN , Regulación de la Expresión Génica , Leucocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/irrigación sanguínea , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Nat Commun ; 13(1): 7029, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396641

RESUMEN

Neutrophil diapedesis is an immediate step following infections and injury and is driven by complex interactions between leukocytes and various components of the blood vessel wall. Here, we show that perivascular mast cells (MC) are key regulators of neutrophil behaviour within the sub-endothelial space of inflamed venules. Using confocal intravital microscopy, we observe directed abluminal neutrophil motility along pericyte processes towards perivascular MCs, a response that created neutrophil extravasation hotspots. Conversely, MC-deficiency and pharmacological or genetic blockade of IL-17A leads to impaired neutrophil sub-endothelial migration and breaching of the pericyte layer. Mechanistically, identifying MCs as a significant cellular source of IL-17A, we establish that MC-derived IL-17A regulates the enrichment of key effector molecules ICAM-1 and CXCL1 in nearby pericytes. Collectively, we identify a novel MC-IL-17A-pericyte axis as modulator of the final steps of neutrophil diapedesis, with potential translational implications for inflammatory disorders driven by increased neutrophil diapedesis.


Asunto(s)
Neutrófilos , Migración Transendotelial y Transepitelial , Neutrófilos/fisiología , Pericitos , Interleucina-17 , Mastocitos
14.
Am J Pathol ; 176(1): 482-95, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20008148

RESUMEN

The venular basement membrane plays a critical role in maintaining the integrity of blood vessels and through its dense and highly organized network of matrix proteins also acts as a formidable barrier to macromolecules and emigrating leukocytes. Leukocytes can however penetrate the venular basement membrane at sites of inflammation, though the associated in vivo mechanisms are poorly understood. Using whole mount immunostained tissues and confocal microscopy, we demonstrate that the venular basement membrane of multiple organs expresses regions of low matrix protein (laminin-511 and type IV collagen) deposition that have been termed low-expression regions (LERs). In the multiple tissues analyzed (eg, cremaster muscle, skin, mesenteric tissue), LERs were directly aligned with gaps between adjacent pericytes and were more prevalent in small venules. As predicted by their permissive nature, LERs acted as "gates" for transmigrating neutrophils in all inflammatory reactions investigated (elicited by leukotriene B(4) [LTB(4)], CXCL1, tumor necrosis factor [TNF]alpha, endotoxin, and ischemia/reperfusion [I/R] injury), and this response was associated with an enhancement of the size of laminin-511 and type IV collagen LERs. Transmigrated neutrophils stained positively for laminins but not type IV collagen, suggesting that different mechanisms exist in remodeling of different basement membrane networks. Collectively the findings provide further insight into characteristics of specialized regions within venular basement membranes that are preferentially used and remodeled by transmigrating neutrophils.


Asunto(s)
Membrana Basal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Inflamación/metabolismo , Inflamación/patología , Especificidad de Órganos , Vénulas/metabolismo , Animales , Membrana Basal/efectos de los fármacos , Capilares/efectos de los fármacos , Capilares/metabolismo , Capilares/patología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Colágeno Tipo IV/metabolismo , Perfilación de la Expresión Génica , Laminina/metabolismo , Lipopolisacáridos/farmacología , Ratones , Modelos Biológicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Factores de Tiempo , Vénulas/efectos de los fármacos , Vénulas/patología
15.
Blood ; 113(24): 6246-57, 2009 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-19211506

RESUMEN

Leukocyte transmigration is mediated by endothelial cell (EC) junctional molecules, but the associated mechanisms remain unclear. Here we investigate how intercellular adhesion molecule-2 (ICAM-2), junctional adhesion molecule-A (JAM-A), and platelet endothelial cell adhesion molecule (PECAM-1) mediate neutrophil transmigration in a stimulus-dependent manner (eg, as induced by interleukin-1beta [IL-1beta] but not tumor necrosis factor-alpha [TNF-alpha]), and demonstrate their ability to act in sequence. Using a cell-transfer technique, transmigration responses of wild-type and TNF-alpha p55/p75 receptor-deficient leukocytes (TNFR(-/-)) through mouse cremasteric venules were quantified by fluorescence intravital microscopy. Whereas wild-type leukocytes showed a normal transmigration response to TNF-alpha in ICAM-2(-/-), JAM-A(-/-), and PECAM-1(-/-) recipient mice, TNFR(-/-) leukocytes exhibited a reduced transmigration response. Hence, when the ability of TNF-alpha to directly stimulate neutrophils is blocked, TNF-alpha-induced neutrophil transmigration is rendered dependent on ICAM-2, JAM-A, and PECAM-1, suggesting that the stimulus-dependent role of these molecules is governed by the target cell being activated. Furthermore, analysis of the site of arrest of neutrophils in inflamed tissues from ICAM-2(-/-), JAM-A(-/-), and PECAM-1(-/-) mice demonstrated that these molecules act sequentially to mediate transmigration. Collectively, the findings provide novel insights into the mechanisms of action of key molecules implicated in leukocyte transmigration.


Asunto(s)
Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Movimiento Celular/fisiología , Endotelio Vascular/metabolismo , Neutrófilos/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Receptores de Superficie Celular/fisiología , Animales , Adhesión Celular , Células Cultivadas , Endotelio Vascular/citología , Técnica del Anticuerpo Fluorescente , Leucocitos/citología , Leucocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculos/citología , Músculos/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/farmacología
17.
Curr Opin Hematol ; 17(1): 9-17, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19864945

RESUMEN

PURPOSE OF REVIEW: As the migration of neutrophils from blood to inflamed tissues is an essential component of innate immunity and a key contributing factor to the pathogenesis of inflammatory disorders, this aspect of leukocyte biology continues to be a highly dynamic field of research. This review summarizes recent findings in this area, focusing on the mechanisms that mediate neutrophil transmigration, an area where significant progress has been made. RECENT FINDINGS: The topics to be covered will include responses that are prerequisite to neutrophil migration through venular walls, such as leukocyte luminal crawling and cellular and molecular changes in leukocytes and endothelial cells (e.g. formation of protrusions) that collectively support leukocyte transendothelial cell migration. Advances in both paracellular and transcellular neutrophil migration through endothelial cells will be discussed, addressing the associated roles and regulation of expression of endothelial cell luminal and junctional adhesion molecules. Beyond the endothelium, migration through the vascular pericyte coverage and basement membrane will be reviewed. SUMMARY: The unquestionable role of neutrophils in the development and progression of inflammatory conditions suggests that a better understanding of the tissue-specific and stimulus-specific mechanisms that mediate this response may identify novel pathways that could be exploited for the development of more specific anti-inflammatory interventions.


Asunto(s)
Movimiento Celular , Células Endoteliales/fisiología , Neutrófilos , Animales , Células Endoteliales/inmunología , Humanos , Inflamación , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/fisiología
18.
Arterioscler Thromb Vasc Biol ; 29(8): 1193-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19498176

RESUMEN

OBJECTIVE: Leukocyte migration through venular walls is a fundamental event during inflammation, but many aspects of this response, including the mechanisms associated with leukocyte migration through the vascular basement membrane (BM) in vivo, are poorly understood. Here we investigated and compared the means by which neutrophils and monocytes migrate through the venular BM. Specifically, as we have previously reported on the existence of neutrophil permissive sites (termed matrix protein low expression regions; LERs) within the venular BM, we have now investigated the role of these sites in monocyte transmigration in vivo. METHODS AND RESULTS: Analysis of CCL2-stimulated mouse cremaster muscles by immunofluorescent staining and confocal microscopy demonstrated that both neutrophils and monocytes use LERs for penetrating venular walls, but independent and distinct mechanisms are used by the 2 cell types. Collectively, (1) neutrophil but not monocyte transmigration led to enlargement of LERs, (2) monocytes showed a greater extent of deformability in migrating through the venular BM, and (3) only extravasated neutrophils were associated with the carriage of laminin fragments. CONCLUSIONS: The findings provide novel insights into mechanisms of leukocyte transmigration by presenting the first in vivo evidence for distinct modes used by neutrophils and monocytes in penetrating the vascular BM.


Asunto(s)
Monocitos/fisiología , Neutrófilos/fisiología , Venas/citología , Animales , Membrana Basal/citología , Membrana Basal/fisiología , Ensayos de Migración Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/farmacología , Masculino , Ratones , Microscopía Confocal , Monocitos/citología , Monocitos/efectos de los fármacos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Venas/fisiología
19.
Arterioscler Thromb Vasc Biol ; 29(10): 1509-15, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19574560

RESUMEN

OBJECTIVE: Junctional adhesion molecule-C (JAM-C) is an adhesion molecule that has multiple roles in inflammation and vascular biology, but many aspects of its functions under pathological conditions are unknown. Here we investigated the role of JAM-C in leukocyte migration in response to ischemia reperfusion (I/R) injury. METHODS AND RESULTS: Pretreatment of mice with soluble JAM-C (sJAM-C), used as a pharmacological blocker of JAM-C-mediated reactions, significantly suppressed leukocyte migration in models of kidney and cremaster muscle I/R injury (39 and 51% inhibition, respectively). Furthermore, in the cremaster muscle model (studied by intravital microscopy), both leukocyte adhesion and transmigration were suppressed in JAM-C-deficient mice (JAM-C(-/-)) and enhanced in mice overexpressing JAM-C in their endothelial cells (ECs). Analysis of JAM-C subcellular expression by immunoelectron microscopy indicated that in I/R-injured tissues, EC JAM-C was redistributed from cytoplasmic vesicles and EC junctional sites to nonjunctional plasma membranes, a response that may account for the role of JAM-C in both leukocyte adhesion and transmigration under conditions of I/R injury. CONCLUSIONS: The findings demonstrate a role for EC JAM-C in mediating leukocyte adhesion and transmigration in response to I/R injury and indicate the existence of a novel regulatory mechanism for redistribution and hence function of EC JAM-C in vivo.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Inmunoglobulinas/fisiología , Leucocitos/fisiología , Daño por Reperfusión/patología , Animales , Adhesión Celular , Moléculas de Adhesión Celular/análisis , Movimiento Celular , Células Endoteliales/metabolismo , Inmunoglobulinas/análisis , Riñón/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/irrigación sanguínea
20.
J Clin Invest ; 130(5): 2301-2318, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31971917

RESUMEN

Increased microvascular permeability to plasma proteins and neutrophil emigration are hallmarks of innate immunity and key features of numerous inflammatory disorders. Although neutrophils can promote microvascular leakage, the impact of vascular permeability on neutrophil trafficking is unknown. Here, through the application of confocal intravital microscopy, we report that vascular permeability-enhancing stimuli caused a significant frequency of neutrophil reverse transendothelial cell migration (rTEM). Furthermore, mice with a selective defect in microvascular permeability enhancement (VEC-Y685F-ki) showed reduced incidence of neutrophil rTEM. Mechanistically, elevated vascular leakage promoted movement of interstitial chemokines into the bloodstream, a response that supported abluminal-to-luminal neutrophil TEM. Through development of an in vivo cell labeling method we provide direct evidence for the systemic dissemination of rTEM neutrophils, and showed them to exhibit an activated phenotype and be capable of trafficking to the lungs where their presence was aligned with regions of vascular injury. Collectively, we demonstrate that increased microvascular leakage reverses the localization of directional cues across venular walls, thus causing neutrophils engaged in diapedesis to reenter the systemic circulation. This cascade of events offers a mechanism to explain how local tissue inflammation and vascular permeability can induce downstream pathological effects in remote organs, most notably in the lungs.


Asunto(s)
Permeabilidad Capilar/inmunología , Microvasos/inmunología , Activación Neutrófila , Neutrófilos/inmunología , Migración Transendotelial y Transepitelial/inmunología , Animales , Permeabilidad Capilar/genética , Masculino , Ratones , Ratones Transgénicos , Microvasos/patología , Neutrófilos/patología , Migración Transendotelial y Transepitelial/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA