Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Immunity ; 57(6): 1225-1242.e6, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38749446

ABSTRACT

Classical monocytes (CMs) are ephemeral myeloid immune cells that circulate in the blood. Emerging evidence suggests that CMs can have distinct ontogeny and originate from either granulocyte-monocyte- or monocyte-dendritic-cell progenitors (GMPs or MDPs). Here, we report surface markers that allowed segregation of murine GMP- and MDP-derived CMs, i.e., GMP-Mo and MDP-Mo, as well as their functional characterization, including fate definition following adoptive cell transfer. GMP-Mo and MDP-Mo yielded an equal increase in homeostatic CM progeny, such as blood-resident non-classical monocytes and gut macrophages; however, these cells differentially seeded various other selected tissues, including the dura mater and lung. Specifically, GMP-Mo and MDP-Mo differentiated into distinct interstitial lung macrophages, linking CM dichotomy to previously reported pulmonary macrophage heterogeneity. Collectively, we provide evidence for the existence of two functionally distinct CM subsets in the mouse that differentially contribute to peripheral tissue macrophage populations in homeostasis and following challenge.


Subject(s)
Cell Differentiation , Macrophages , Monocytes , Animals , Monocytes/immunology , Monocytes/cytology , Mice , Cell Differentiation/immunology , Macrophages/immunology , Macrophages/metabolism , Lung/cytology , Lung/immunology , Homeostasis , Mice, Inbred C57BL , Dendritic Cells/immunology , Cell Lineage , Adoptive Transfer
3.
Immunity ; 49(6): 1062-1076.e6, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30446388

ABSTRACT

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.


Subject(s)
Chemokine CXCL1 , Chemokine CXCL2 , Duffy Blood-Group System , Neutrophils , Receptors, Cell Surface , Transendothelial and Transepithelial Migration , Animals , Abdominal Muscles/drug effects , Abdominal Muscles/immunology , Abdominal Muscles/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Chemokine CXCL1/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Chemokine CXCL2/metabolism , Duffy Blood-Group System/genetics , Duffy Blood-Group System/immunology , Duffy Blood-Group System/metabolism , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Intercellular Junctions/drug effects , Intercellular Junctions/immunology , Intercellular Junctions/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/genetics , Transendothelial and Transepithelial Migration/immunology , Tumor Necrosis Factor-alpha/pharmacology
4.
Immunity ; 47(5): 805-807, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166580

ABSTRACT

mTORC2 is a metabolic regulatory complex activated by PI3K. In this issue of Immunity, Kishore et al. (2017) demonstrate a specialized role for this complex in the migration of regulatory T (Treg) cells to sites of inflammation rather than their differentiation and survival.


Subject(s)
Glycolysis , T-Lymphocytes, Regulatory , Cell Movement , Glucokinase , Mechanistic Target of Rapamycin Complex 2
5.
Trends Immunol ; 42(8): 706-722, 2021 08.
Article in English | MEDLINE | ID: mdl-34266767

ABSTRACT

The integrin LFA-1 is crucial for T cell entry into mammalian lymph nodes and tissues, and for promoting interactions with antigen-presenting cells (APCs). However, it is increasingly evident that LFA-1 has additional key roles beyond the mere support of adhesion between T cells, the endothelium, and/or APCs. These include roles in homotypic T cell-T cell (T-T) communication, the induction of intracellular complement activity underlying Th1 effector cell polarization, and the support of long-lasting T cell memory. Here, we briefly summarize current knowledge of LFA-1 biology, discuss novel cytoskeletal regulators of LFA-1 functions, and review new aspects of LFA-1 mechanobiology that are relevant to its function in immunological synapses and in specific pathologies arising from LFA-1 dysregulation.


Subject(s)
Intercellular Adhesion Molecule-1 , Lymphocyte Function-Associated Antigen-1 , Animals , Antigen-Presenting Cells , Cell Differentiation , Th1 Cells
6.
Nat Immunol ; 13(1): 67-76, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138716

ABSTRACT

Chemokines presented by the endothelium are critical for integrin-dependent adhesion and transendothelial migration of naive and memory lymphocytes. Here we found that effector lymphocytes of the type 1 helper T cell (T(H)1 cell) and type 1 cytotoxic T cell (T(C)1 cell) subtypes expressed adhesive integrins that bypassed chemokine signals and established firm arrests on variably inflamed endothelial barriers. Nevertheless, the transendothelial migration of these lymphocytes strictly depended on signals from guanine nucleotide-binding proteins of the G(i) type and was promoted by multiple endothelium-derived inflammatory chemokines, even without outer endothelial surface exposure. Instead, transendothelial migration-promoting endothelial chemokines were stored in vesicles docked on actin fibers beneath the plasma membranes and were locally released within tight lymphocyte-endothelial synapses. Thus, effector T lymphocytes can cross inflamed barriers through contact-guided consumption of intraendothelial chemokines without surface-deposited chemokines or extraendothelial chemokine gradients.


Subject(s)
Chemokines/metabolism , Endothelial Cells/metabolism , Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Transport Vesicles/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Endothelial Cells/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Hyaluronan Receptors/metabolism , Integrins/metabolism , Lymphocytes/metabolism , Lymphocytes/ultrastructure , Mice , Receptors, CCR2/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/ultrastructure , Tumor Necrosis Factor-alpha/pharmacology , Vasculitis/immunology , Vasculitis/metabolism
7.
Immunity ; 41(5): 694-707, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25517612

ABSTRACT

Leukocyte migration through activated venular walls is a fundamental immune response that is prerequisite to the entry of effector cells such as neutrophils, monocytes, and effector T cells to sites of infection, injury, and stress within the interstitium. Stimulation of leukocytes is instrumental in this process with enhanced temporally controlled leukocyte adhesiveness and shape-changes promoting leukocyte attachment to the inner wall of blood vessels under hydrodynamic forces. This initiates polarized motility of leukocytes within and through venular walls and transient barrier disruption facilitated sequentially by stimulated vascular cells, i.e., endothelial cells and their associated pericytes. Perivascular cells such as macrophages and mast cells that act as tissue inflammatory sentinels can also directly and indirectly regulate the exit of leukocytes from the vascular lumen. In this review, we discuss current knowledge and open questions regarding the mechanisms involved in the interactions of different effector leukocytes with peripheral vessels in extralymphoid organs.


Subject(s)
Blood Vessels/immunology , Endothelium, Vascular/immunology , Inflammation/immunology , Leukocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Cell Adhesion/immunology , Humans , Integrins/metabolism , Macrophages/immunology , Mast Cells/immunology , Neutrophils/immunology , T-Lymphocytes/immunology
8.
Biophys J ; 120(18): 4002-4012, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34411577

ABSTRACT

Leukocyte microvilli are elastic actin-rich projections implicated in rapid sensing and penetration across glycocalyx barriers. Microvilli are critical for the capture and arrest of flowing lymphocytes by high endothelial venules, the main lymph node portal vessels. T lymphocyte arrest involves subsecond activation of the integrin LFA-1 by the G-protein-coupled receptor CCR7 and its endothelial-displayed ligands, the chemokines CCL21 and CCL19. The topographical distribution of CCR7 and of LFA-1 in relation to lymphocyte microvilli has never been elucidated. We applied the recently developed microvillar cartography imaging technique to determine the topographical distribution of CCR7 and LFA-1 with respect to microvilli on peripheral blood T lymphocytes. We found that CCR7 is clustered on the tips of T cell microvilli. The vast majority of LFA-1 molecules were found on the cell body, likely assembled in macroclusters, but a subset of LFA-1, 5% of the total, were found scattered within 20 nm from the CCR7 clusters, implicating these LFA-1 molecules as targets for inside-out activation signals transmitted within a fraction of a second by chemokine-bound CCR7. Indeed, RhoA, the key GTPase involved in rapid LFA-1 affinity triggering by CCR7, was also found to be clustered near CCR7. In addition, we observed that the tyrosine kinase JAK2 controls CCR7-mediated LFA-1 affinity triggering and is also highly enriched on tips of microvilli. We propose that tips of lymphocyte microvilli are novel signalosomes for subsecond CCR7-mediated inside-out signaling to neighboring LFA-1 molecules, a critical checkpoint in LFA-1-mediated lymphocyte arrest on high endothelial venules.


Subject(s)
Chemokine CCL21 , Lymphocyte Function-Associated Antigen-1 , Lymphocytes , Microvilli , Receptors, CCR7
10.
Blood ; 131(12): 1337-1349, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29352038

ABSTRACT

Chronic lymphocytic leukemia (CLL) outgrowth depends on signals from the microenvironment. We have previously found that in vitro reconstitution of this microenvironment induces specific variant isoforms of the adhesion molecule CD44, which confer human CLL with high affinity to hyaluronan (HA). Here, we determined the in vivo contribution of standard CD44 and its variants to leukemic B-cell homing and proliferation in Tcl1 transgenic mice with a B-cell-specific CD44 deficiency. In these mice, leukemia onset was delayed and leukemic infiltration of spleen, liver, and lungs, but not of bone marrow, was decreased. Competitive transplantation revealed that CLL homing to spleen and bone marrow required functional CD44. Notably, enrichment of CD44v6 variants particularly in spleen enhanced CLL engraftment and proliferation, along with increased HA binding. We recapitulated CD44v6 induction in the human disease and revealed the involvement of MAPK and NF-κB signaling upon CD40 ligand and B-cell receptor stimulation by in vitro inhibition experiments and chromatin immunoprecipitation assays. The investigation of downstream signaling after CD44v6-HA engagement uncovered the activation of extracellular signal-regulated kinase and p65. Consequently, anti-CD44v6 treatment reduced leukemic cell proliferation in vitro in human and mouse, confirming the general nature of the findings. In summary, we propose a CD44-NF-κB-CD44v6 circuit in CLL, allowing tumor cells to gain HA binding capacity and supporting their proliferation.


Subject(s)
Cell Proliferation , Hyaluronan Receptors/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Tumor Microenvironment , Animals , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/genetics , Hyaluronic Acid/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Spleen/metabolism , Spleen/pathology
11.
Trends Immunol ; 38(8): 606-615, 2017 08.
Article in English | MEDLINE | ID: mdl-28559148

ABSTRACT

Leukocyte transendothelial migration (TEM) takes place across micron-wide gaps in specific post-capillary venules generated by the transmigrating leukocyte. Because endothelial cells contain a dense cytoskeletal network, transmigrating leukocytes must overcome these mechanical barriers as they squeeze their nuclei through endothelial gaps and pores. Recent findings suggest that endothelial cells are not a passive barrier, and upon engagement by transmigrating leukocytes trigger extensive dynamic modifications of their actin cytoskeleton. Unexpectedly, endothelial contractility functions as a restrictor of endothelial gap enlargement rather than as a facilitator of gap formation as was previously suggested. In this review we discuss current knowledge regarding how accurately timed endothelial actin-remodeling events are triggered by squeezing leukocytes and coordinate leukocyte TEM while preserving blood vessel integrity.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelium, Vascular/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Transendothelial and Transepithelial Migration , Animals , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Endothelium, Vascular/cytology , Humans , Junctional Adhesion Molecules/metabolism
12.
Immunity ; 33(5): 654-6, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21094462

ABSTRACT

In this issue of Immunity, Bao et al. (2010) provide in vivo evidence that heparan sulfate glycosaminoglycans (GAGs) are indispensable for immobilization and function of major chemokines required for leukocyte adhesion to and crossing through blood and lymphatic vessels.

13.
Proc Natl Acad Sci U S A ; 113(40): E5916-E5924, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647916

ABSTRACT

Leukocyte microvilli are flexible projections enriched with adhesion molecules. The role of these cellular projections in the ability of T cells to probe antigen-presenting cells has been elusive. In this study, we probe the spatial relation of microvilli and T-cell receptors (TCRs), the major molecules responsible for antigen recognition on the T-cell membrane. To this end, an effective and robust methodology for mapping membrane protein distribution in relation to the 3D surface structure of cells is introduced, based on two complementary superresolution microscopies. Strikingly, TCRs are found to be highly localized on microvilli, in both peripheral blood human T cells and differentiated effector T cells, and are barely found on the cell body. This is a decisive demonstration that different types of T cells universally localize their TCRs to microvilli, immediately pointing to these surface projections as effective sensors for antigenic moieties. This finding also suggests how previously reported membrane clusters might form, with microvilli serving as anchors for specific T-cell surface molecules.


Subject(s)
Microscopy/methods , Microvilli/metabolism , Receptors, Antigen, T-Cell/metabolism , Antigens, CD/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Humans , Imaging, Three-Dimensional , L-Selectin/metabolism , Lymph Nodes/cytology , Lymph Nodes/metabolism , Membrane Proteins/metabolism , Microvilli/drug effects , Microvilli/ultrastructure , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/ultrastructure , Thiazolidines/pharmacology
14.
Immunity ; 30(3): 384-96, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19268609

ABSTRACT

Endothelial chemokines are instrumental for integrin-mediated lymphocyte adhesion and transendothelial migration (TEM). By dissecting how chemokines trigger lymphocyte integrins to support shear-resistant motility on and across cytokine-stimulated endothelial barriers, we found a critical role for high-affinity (HA) LFA-1 integrin in lymphocyte crawling on activated endothelium. Endothelial-presented chemokines triggered HA-LFA-1 and adhesive filopodia at numerous submicron dots scattered underneath crawling lymphocytes. Shear forces applied to endothelial-bound lymphocytes dramatically enhanced filopodia density underneath crawling lymphocytes. A fraction of the adhesive filopodia invaded the endothelial cells prior to and during TEM and extended large subluminal leading edge containing dots of HA-LFA-1 occupied by subluminal ICAM-1. Memory T cells generated more frequent invasive filopodia and transmigrated more rapidly than their naive counterparts. We propose that shear forces exerted on HA-LFA-1 trigger adhesive and invasive filopodia at apical endothelial surfaces and thereby promote lymphocyte crawling and probing for TEM sites.


Subject(s)
Cell Movement , Chemokines/immunology , Endothelium, Vascular/immunology , Lymphocyte Function-Associated Antigen-1/immunology , T-Lymphocytes/immunology , Cells, Cultured , Humans , Intercellular Adhesion Molecule-1/immunology
15.
FASEB J ; 30(5): 1767-78, 2016 05.
Article in English | MEDLINE | ID: mdl-26823454

ABSTRACT

The pulmonary vasculature constitutively expresses the integrin lymphocyte function-associated antigen-1 ligands intercellular adhesion molecule (ICAM)-1 and -2. In this study, effector T cells were temporarily entrapped by the lung vasculature on their way to inflamed lymph nodes, and this entrapment was strongly reduced in ICAM-1 and -2 double-deficient mice (79 and 86% reduction for CD8(+) and CD4(+) effectors, respectively, compared with wild-type mice). Although the pulmonary vasculature has been suggested to be masked by the heparan sulfate-containing glycocalyx, which is susceptible to heparanase-mediated shedding, lung and lymphocyte heparanase have been found to be unnecessary for this entrapment. Systemic LPS induced rapid neutrophil entrapment in the lung vasculature, but in contrast to T-cell entrapment, this sequestration was ICAM-1, ICAM-2, and heparanase independent. Furthermore, neutrophil migration into the bronchoalveolar space induced by LPS inhalation and LPS-induced leakage of red blood cells into this space were not dependent on lung ICAMs or heparanase activity. Nevertheless, heparanase was critical for neutrophil accumulation in smoke-exposed lungs. Our results indicate that, whereas T cells use ICAM-1 and -2 for temporary pulmonary entrapment, neutrophils get sequestered and extravasate into inflamed lungs independent of ICAMs. This is the first demonstration that the pulmonary vasculature is differentially recognized by T cells and neutrophils.-Petrovich, E., Feigelson, S. W., Stoler-Barak, L., Hatzav, M., Solomon, A., Bar-Shai, A., Ilan, N., Li, J.-P., Engelhardt, B., Vlodavsky, I., Alon, R. Lung ICAM-1 and ICAM-2 support spontaneous intravascular effector lymphocyte entrapment but are not required for neutrophil entrapment or emigration inside endotoxin-inflamed lungs.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Inflammation/chemically induced , Intercellular Adhesion Molecule-1/metabolism , Lung Diseases/chemically induced , Lymphocytes/physiology , Neutrophils/physiology , Animals , Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Cell Movement , Endotoxins/toxicity , Gene Expression Regulation/physiology , Glucuronidase/metabolism , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/genetics , Lung/blood supply , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice
16.
FASEB J ; 29(5): 2010-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25634957

ABSTRACT

Heparanase, the exclusive mammalian heparan sulfate-degrading enzyme, has been suggested to be utilized by leukocytes to penetrate through the dense basement membranes surrounding blood venules. Despite its established role in tumor cell invasion, heparanase function in leukocyte extravasation has never been demonstrated. We found that TH1/TC1-type effector T cells are highly enriched for this enzyme, with a 3.6-fold higher heparanase mRNA expression compared with naive lymphocytes. Using adoptive transfer of wild-type and heparanase-deficient effector T cells into inflamed mice, we show that T-cell heparanase was not required for extravasation inside inflamed lymph nodes or skin. Leukocyte extravasation through acute inflamed skin vessels was also heparanase independent. Furthermore, neutrophils emigrated to the inflamed peritoneal cavity independently of heparanase expression on either the leukocytes or on the endothelial and mesothelial barriers, and overexpression of the enzyme on neutrophils did not facilitate their emigration. However, heparanase absence significantly reduced monocyte emigration into the inflamed peritoneal cavity. These results collectively suggest that neither leukocyte nor endothelial heparanase is required for T-cell and neutrophil extravasation through inflamed vascular barriers, whereas this enzyme is required for optimal monocyte recruitment to inflamed peritoneum.


Subject(s)
Endothelium, Vascular/immunology , Glucuronidase/physiology , Inflammation/immunology , Neutrophils/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Female , Flow Cytometry , Inflammation/enzymology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/cytology , Neutrophils/enzymology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/enzymology , Skin/pathology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology
17.
Blood ; 122(15): 2609-17, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23980064

ABSTRACT

Kindlin-3 is an integrin-binding focal adhesion adaptor absent in patients with leukocyte and platelet adhesion deficiency syndrome and is critical for firm integrin-dependent leukocyte adhesion. The role of this adaptor in leukocyte diapedesis has never been investigated. In the present study, the functions of Kindlin-3 in this process were investigated in effector T lymphocytes trafficking to various lymphoid and nonlymphoid tissues. In vitro, Kindlin-3-deficient T cells displayed severely impaired lymphocyte function antigen-1-dependent lymphocyte adhesion but partially conserved very late antigen-4 adhesiveness. In vivo, the number of adoptively transferred Kindlin-3-deficient T effectors was dramatically elevated in the circulating pool compared with normal effectors, and the Kindlin-3 mutant effectors failed to enter inflamed skin lesions. The frequency of Kindlin-3-deficient T effectors arrested on vessel walls within inflamed skin-draining lymph nodes was also reduced. Strikingly, however, Kindlin-3-deficient effector T cells accumulated inside these vessels at significantly higher numbers than their wild-type lymphocyte counterparts and successfully extravasated into inflamed lymph nodes. Nevertheless, on entering these organs, the interstitial motility of these lymphocytes was impaired. This is the first in vivo demonstration that Kindlin-3-stabilized integrin adhesions, although essential for lymphocyte arrest on blood vessels and interstitial motility, are not obligatory for leukocyte diapedesis.


Subject(s)
Cytoskeletal Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Vasculitis/immunology , Adoptive Transfer , Animals , Cell Adhesion/immunology , Cell Movement/immunology , Cytoskeletal Proteins/deficiency , Dermatitis/immunology , Dermatitis/pathology , Humans , Integrin alpha4beta1/immunology , Lymphadenitis/immunology , Lymphadenitis/pathology , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred C57BL , Vasculitis/pathology
18.
Curr Opin Cell Biol ; 20(5): 525-32, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18499427

ABSTRACT

The arrest of rolling leukocytes on various target vascular beds is mediated by specialized leukocyte integrins and their endothelial immunoglobulin superfamily (IgSF) ligands. These integrins are kept in largely inactive states and undergo in situ activation upon leukocyte-endothelial contact by both biochemical and mechanical signals from flow-derived shear forces. In vivo and in vitro studies suggest that leukocyte integrin activation involves conformational alterations through inside-out signaling followed by ligand-induced rearrangements accelerated by external forces. This activation process takes place within fractions of seconds by in situ signals transduced to the rolling leukocyte as it encounters specialized endothelial-displayed chemoattractants, collectively termed arrest chemokines. In neutrophils, selectin rolling engagements trigger intermediate affinity integrins to support reversible adhesions before chemokine-triggered arrest. Different leukocyte subsets appear to use different modalities of integrin activation during rolling and arrest at distinct endothelial sites.


Subject(s)
Endothelial Cells/metabolism , Integrins/metabolism , Leukocyte Rolling/physiology , Lymphocyte Activation/physiology , Stress, Mechanical , Chemokines/metabolism , Cytoskeleton/metabolism , GTP Phosphohydrolases/metabolism , Humans , Integrins/chemistry , Ligands , Models, Molecular , Protein Conformation , Receptors, G-Protein-Coupled/metabolism , Selectins/metabolism , Shear Strength , Signal Transduction/physiology
19.
Int Immunol ; 26(6): 315-24, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24402310

ABSTRACT

Leukocyte diapedesis is a chemotactic multistep process that requires optimal chemoattractant presentation by the endothelial barrier. Recent studies have described a critical role for heparan sulfate glycosaminoglycans (HSGAGs) in the presentation and functions of chemokines essential for lymphocyte interactions with the lymph node vasculature. We wished to test whether HS expression by a prototypic endothelial cell type, i.e. human umbilical vein endothelial cells (HUVECs), is critical for their ability to support neutrophil and lymphocyte adhesion and transendothelial migration (TEM) under shear flow. We found that HUVECs deposit HS GAGs mainly at their basolateral compartments in both their resting and inflamed states. We next inactivated the key enzyme involved in HS biosynthesis, exostosin-1 (Ext1). Silencing Ext1 resulted in a complete loss of HS biosynthesis; nonetheless, TNF-α and IL-1ß stimulation of key adhesion molecules and inflammatory chemokines necessary for neutrophil or lymphocyte adhesion and TEM remained intact. Ext1 silencing reduced neutrophil arrest and markedly impaired TEM, consistent with a role of basolateral HS GAGs in directing neutrophil crossing of inflamed endothelial barriers. Strikingly, however, the TEM of effector T cells across identically Ext1-silenced HUVECs remained normal. Importantly, the biosynthesis of the main promigratory chemokines for effector T cells and neutrophils, respectively, CCL2 and CXCL1, and their vesicle distributions were also Ext1 independent. These results suggest that transmigrating neutrophils must respond to chemokines transiently presented by apical and basolateral endothelial HS GAGs. In contrast, effector T cells can integrate chemotactic TEM signals directly from intra-endothelial chemokine stores rather than from externally deposited chemokines.


Subject(s)
Endothelium, Vascular/metabolism , Heparin/analogs & derivatives , N-Acetylglucosaminyltransferases/metabolism , Neutrophils/immunology , Proteoglycans/metabolism , T-Lymphocytes/immunology , Cell Line , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Chemotaxis , Heparin/metabolism , Humans , Inflammation/immunology , Interleukin-1beta/metabolism , N-Acetylglucosaminyltransferases/genetics , RNA, Small Interfering/genetics , Transendothelial and Transepithelial Migration/genetics , Tumor Necrosis Factor-alpha
20.
Respiration ; 90(1): 74-84, 2015.
Article in English | MEDLINE | ID: mdl-26068137

ABSTRACT

BACKGROUND: Acute lung inflammation can be monitored by various biochemical readouts of bronchoalveolar lavage fluid (BALF). OBJECTIVE: To analyze the BALF content of ultrafine particles (UFP; <100 nm) as an inflammatory biomarker in early diagnosis of acute and chronic lung diseases. METHODS: Mice were exposed to different stress conditions and inflammatory insults (acute lipopolysaccharide inhalation, tobacco smoke and lethal dose of total body irradiation, i.e. 950 rad). After centrifugation, the cellular pellet was assessed while cytokines and ultrafine particles were measured in the soluble fraction of the BALF. RESULTS: A characteristic UFP distribution with a D50 (i.e. the dimension of the 50th UFP percentile) was shared by all tested mouse strains in the BALF of resting lungs. All tested inflammatory insults similarly shifted this size distribution, resulting in a unique UFP fingerprint with an averaged D50 of 58.6 nm, compared with the mean UFP D50 of 23.7 nm for resting BALF (p < 0.0001). This UFP profile was highly reproducible and independent of the intensity or duration of the inflammatory trigger. It returned to baseline after resolution of the inflammation. Neither total body irradiation nor induction of acute cough induced this fingerprint. CONCLUSIONS: The UFP fingerprint in the BALF of resting and inflamed lungs can serve as a binary biomarker of healthy and acutely inflamed lungs. This marker can be used as a novel readout for the onset of inflammatory lung diseases and for complete lung recovery from different insults.


Subject(s)
Bronchoalveolar Lavage Fluid , Lipopolysaccharides/pharmacology , Lung , Particulate Matter/analysis , Radiation Pneumonitis , Smoke , Animals , Inflammation , Inhalation Exposure , Lung/drug effects , Lung/radiation effects , Mice , Particle Size , Pneumonia , Radiation , Nicotiana
SELECTION OF CITATIONS
SEARCH DETAIL