Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Front Immunol ; 12: 663886, 2021.
Article in English | MEDLINE | ID: mdl-33995392

ABSTRACT

Neutrophils are essential to protect the host against invading pathogens but can promote disease progression in sickle cell disease (SCD) by becoming adherent to inflamed microvascular networks in peripheral tissue throughout the body. During the inflammatory response, leukocytes extravasate from the bloodstream using selectin adhesion molecules and migrate to sites of tissue insult through activation of integrins that are essential for combating pathogens. However, during vaso-occlusion associated with SCD, neutrophils are activated during tethering and rolling on selectins upregulated on activated endothelium that line blood vessels. Recently, we reported that recognition of sLex on L-selectin by E-selectin during neutrophil rolling initiates shear force resistant catch-bonds that facilitate tethering to endothelium and activation of integrin bond clusters that anchor cells to the vessel wall. Evidence indicates that blocking this important signaling cascade prevents the congestion and ischemia in microvasculature that occurs from neutrophil capture of sickled red blood cells, which are normally deformable ellipses that flow easily through small blood vessels. Two recently completed clinical trials of therapies targeting selectins and their effect on neutrophil activation in small blood vessels reveal the importance of mechanoregulation that in health is an immune adaption facilitating rapid and proportional leukocyte adhesion, while sustaining tissue perfusion. We provide a timely perspective on the mechanism underlying vaso-occlusive crisis (VOC) with a focus on new drugs that target selectin mediated integrin adhesive bond formation.


Subject(s)
Anemia, Sickle Cell/complications , Cell Adhesion/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Animals , Blood Platelets/drug effects , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Adhesion Molecules/metabolism , Disease Management , Disease Susceptibility , Endothelium/metabolism , Humans , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Neutrophil Activation , Neutrophils/immunology , Selectins/metabolism
2.
J Immunol ; 205(12): 3300-3310, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33199537

ABSTRACT

Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1ß production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.


Subject(s)
Colchicine/pharmacology , Gout , Leukocyte Rolling , Neutrophil Infiltration/drug effects , Neutrophils , Rho Guanine Nucleotide Exchange Factors/immunology , Actins/genetics , Actins/immunology , Animals , Disease Models, Animal , Gout/drug therapy , Gout/genetics , Gout/immunology , Gout/pathology , Leukocyte Rolling/drug effects , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Myosin Light Chains , Neutrophils/immunology , Neutrophils/pathology , Rho Guanine Nucleotide Exchange Factors/genetics
3.
Methods Mol Biol ; 2080: 19-25, 2020.
Article in English | MEDLINE | ID: mdl-31745867

ABSTRACT

The exit of leukocytes from the bloodstream into inflamed sites involves a sequence of interactions with vascular endothelial cells, in which leukocytes, moving rapidly in flowing blood, first tether and roll on the endothelial surface before arresting and then transmigrating across the endothelial barrier. Examining the mechanisms of these interactions in human systems has involved the use of in vitro flow chamber assays, using a variety of cells and immobilized molecules as adhesive substrata. Here we describe how to perform these assays using human umbilical vein endothelial cells and human leukocytes.


Subject(s)
Cell Communication , Endothelial Cells/physiology , Hemodynamics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Leukocytes/physiology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Cell Adhesion , Cells, Cultured , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Transendothelial and Transepithelial Migration
4.
Sci Rep ; 8(1): 5317, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593228

ABSTRACT

Chronic kidney disease (CKD) is associated with excessive mortality from cardiovascular disease (CVD). Endothelial dysfunction, an early manifestation of CVD, is consistently observed in CKD patients and might be linked to structural defects of the microcirculation including microvascular rarefaction. However, patterns of microvascular rarefaction in CKD and their relation to functional deficits in perfusion and oxygen delivery are currently unknown. In this in-vivo microscopy study of the cremaster muscle microcirculation in BALB/c mice with moderate to severe uremia, we show in two experimental models (adenine feeding or subtotal nephrectomy), that serum urea levels associate incrementally with a distinct microangiopathy. Structural changes were characterized by a heterogeneous pattern of focal microvascular rarefaction with loss of coherent microvascular networks resulting in large avascular areas. Corresponding microvascular dysfunction was evident by significantly diminished blood flow velocity, vascular tone, and oxygen uptake. Microvascular rarefaction in the cremaster muscle paralleled rarefaction in the myocardium, which was accompanied by a decrease in transcription levels not only of the transcriptional regulator HIF-1α, but also of its target genes Angpt-2, TIE-1 and TIE-2, Flkt-1 and MMP-9, indicating an impaired hypoxia-driven angiogenesis. Thus, experimental uremia in mice associates with systemic microvascular disease with rarefaction, tissue hypoxia and dysfunctional angiogenesis.


Subject(s)
Hypoxia/etiology , Hypoxia/metabolism , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Renal Insufficiency, Chronic/complications , Vascular Diseases/etiology , Vascular Diseases/metabolism , Abdominal Muscles/blood supply , Animals , Biomarkers , Blood Flow Velocity , Blood Pressure , Coronary Vessels , Disease Models, Animal , Leukocyte Rolling/immunology , Male , Mice , Microcirculation , Microvascular Rarefaction , Myocardium , Neovascularization, Pathologic/physiopathology , Oxygen/metabolism , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/metabolism , Severity of Illness Index , Uremia/diagnosis , Uremia/etiology , Vascular Diseases/physiopathology
5.
J Leukoc Biol ; 102(3): 699-709, 2017 09.
Article in English | MEDLINE | ID: mdl-28619950

ABSTRACT

Neutrophils are the first cells arriving at sites of tissue injury or infection to combat invading pathogens. Successful neutrophil recruitment to sites of inflammation highly depends on specific molecular mechanisms, fine-tuning the received information into signaling pathways and converting them into well-described recruitment steps. This review highlights the impact of vascular flow conditions on neutrophil recruitment and the multitude of mechanisms developed to enable this sophisticated process under wall shear stress conditions. The recruitment process underlies a complex interplay between adhesion and signaling molecules, as well as chemokines, in which neutrophils developed specific mechanisms to travel to sites of lesion in low and high shear stress conditions. Rolling, as the first step in the recruitment process, highly depends on endothelial selectins and their ligands on neutrophils, inducting of intracellular signaling and subsequently activating ß2 integrins, enabling adhesion and postadhesion events. In addition, subcellular structures, such as microvilli, tethers, and slings allow the cell to arrest, even under high wall shear stress. Thereby, microvilli that are pulled out from the cell body form tethers that develop into slings upon their detachment from the substrate. In addition to the above-described primary capture, secondary capture of neutrophils via neutrophil-neutrophil or neutrophil-platelet interaction promotes the process of neutrophil recruitment to sites of lesion. Thus, precise mechanisms based on a complex molecular interplay, subcellular structures, and cell-cell interactions turn the delicate process of neutrophil trafficking during flow into a robust response allowing effective neutrophil accumulation at sites of injury.


Subject(s)
Blood Platelets/immunology , Blood Vessels/immunology , Cell Communication/immunology , Neutrophils/immunology , Shear Strength , Stress, Physiological/immunology , CD18 Antigens/immunology , Leukocyte Rolling/immunology
6.
Eur J Immunol ; 46(9): 2187-203, 2016 09.
Article in English | MEDLINE | ID: mdl-27338806

ABSTRACT

Although CD8(+) T cells have been implied in the pathogenesis of multiple sclerosis (MS), the molecular mechanisms mediating CD8(+) T-cell migration across the blood-brain barrier (BBB) into the central nervous system (CNS) are ill defined. Using in vitro live cell imaging, we directly compared the multistep extravasation of activated CD4(+) and CD8(+) T cells across primary mouse brain microvascular endothelial cells (pMBMECs) as a model for the BBB under physiological flow. Significantly higher numbers of CD8(+) than CD4(+) T cells arrested on pMBMECs under noninflammatory and inflammatory conditions. While CD4(+) T cells polarized and crawled prior to their diapedesis, the majority of CD8(+) T cells stalled and readily crossed the pMBMEC monolayer preferentially via a transcellular route. T-cell arrest and crawling were independent of G-protein-coupled receptor signaling. Rather, absence of endothelial ICAM-1 and ICAM-2 abolished increased arrest of CD8(+) over CD4(+) T cells and abrogated T-cell crawling, leading to the efficient reduction of CD4(+) , but to a lesser degree of CD8(+) , T-cell diapedesis across ICAM-1(null) /ICAM-2(-/-) pMBMECs. Thus, cellular and molecular mechanisms mediating the multistep extravasation of activated CD8(+) T cells across the BBB are distinguishable from those involved for CD4(+) T cells.


Subject(s)
Blood-Brain Barrier/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Transcellular Cell Migration/immunology , Animals , Biomarkers , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Leukocyte Rolling/immunology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism
7.
J Biol Chem ; 291(3): 1441-7, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26631722

ABSTRACT

In humans and mice, megakaryocytes/platelets and endothelial cells constitutively synthesize P-selectin and mobilize it to the plasma membrane to mediate leukocyte rolling during inflammation. TNF-α, interleukin 1ß, and LPS markedly increase P-selectin mRNA in mice but decrease P-selectin mRNA in humans. Transgenic mice bearing the entire human SELP gene recapitulate basal and inducible expression of human P-selectin and reveal human-specific differences in P-selectin function. Differences in the human SELP and murine Selp promoters account for divergent expression in vitro, but their significance in vivo is not known. Here we generated knockin mice that replace the 1.4-kb proximal Selp promoter with the corresponding SELP sequence (Selp(KI)). Selp(KI) (/) (KI) mice constitutively expressed more P-selectin on platelets and more P-selectin mRNA in tissues but only slightly increased P-selectin mRNA after injection of TNF-α or LPS. Consistent with higher basal expression, leukocytes rolled more slowly on P-selectin in trauma-stimulated venules of Selp(KI) (/) (KI) mice. However, TNF-α did not further reduce P-selectin-dependent rolling velocities. Blunted up-regulation of P-selectin mRNA during contact hypersensitivity reduced P-selectin-dependent inflammation in Selp(KI) (/-) mice. Higher basal P-selectin in Selp(KI) (/) (KI) mice compensated for this defect. Therefore, divergent sequences in a short promoter mediate most of the functionally significant differences in expression of human and murine P-selectin in vivo.


Subject(s)
Gene Expression Regulation , P-Selectin/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Crosses, Genetic , Dermatitis, Contact/immunology , Dermatitis, Contact/metabolism , Gene Expression Regulation/drug effects , Humans , Leukocyte Rolling/drug effects , Leukocyte Rolling/immunology , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , P-Selectin/chemistry , P-Selectin/genetics , Promoter Regions, Genetic/drug effects , RNA, Messenger/metabolism , Species Specificity , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/metabolism , Venules/drug effects , Venules/immunology
8.
J Immunol ; 196(3): 1305-1316, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26700769

ABSTRACT

Endothelial E- and P-selectins mediate lymphocyte trafficking in inflammatory processes by interacting with lymphocyte selectin ligands. These are differentially expressed among different T cell subsets and function alone or in cooperation to mediate T cell adhesion. In this study, we characterize the expression and functionality of E-selectin ligands in Th type 17 lymphocytes (Th17 cells) and report that CD43 functions as a Th17 cell E-selectin ligand in vitro that mediates Th17 cell rolling on the vascular endothelium and recruitment in vivo. We demonstrate Th17 cells express CD44, P-selectin glycoprotein ligand (PSGL)-1, and CD43. Few PSGL-1(-/-)CD43(-/-) Th17 cells accumulated on E-selectin under shear flow conditions compared with wild-type cells. CD43(-/-) Th17 cell accumulation on E-selectin was impaired as compared with wild-type and PSGL-1(-/-), and similar to that observed for PSGL-1(-/-)CD43(-/-) Th17 cells, indicating that CD43 alone is a dominant ligand for E-selectin. Notably, this finding is Th17 cell subset specific because CD43 requires cooperation with PSGL-1 in Th1 cells for binding to E-selectin. In vivo, Th17 cell recruitment into the air pouch was reduced in CD43(-/-) mice in response to CCL20 or TNF-α, and intravital microscopy studies demonstrated that CD43(-/-) Th17 cells had impaired rolling on TNF-α-treated microvessels. Furthermore, CD43(-/-) mice were protected from experimental autoimmune encephalomyelitis and had impaired recruitment of Th17 cells in the spinal cord. Our findings demonstrate that CD43 is a major E-selectin ligand in Th17 cells that functions independent of PSGL-1, and they suggest that CD43 may hold promise as a therapeutic target to modulate Th17 cell recruitment.


Subject(s)
E-Selectin/immunology , Inflammation/immunology , Leukocyte Rolling/immunology , Leukosialin/immunology , Th17 Cells/immunology , Animals , Blotting, Western , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelium, Vascular/immunology , Flow Cytometry , Immunoprecipitation , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
J Immunol ; 195(8): 3880-9, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26355151

ABSTRACT

A2A adenosine receptor (A2AAR) signaling negatively regulates inflammatory responses in many disease models, but the detailed mechanisms remain unclear. We used the selective A2AAR agonist, ATL313, to examine how A2AAR signaling affects human and murine neutrophil adhesion under flow. Treating neutrophils with ATL313 inhibited selectin-induced, ß2 integrin-dependent slow rolling and chemokine-induced, ß2 integrin-dependent arrest on ICAM-1. ATL313 inhibited selectin-induced ß2 integrin extension, which supports slow rolling, and chemokine-induced hybrid domain "swing-out," which supports arrest. Furthermore, ATL313 inhibited integrin outside-in signaling as revealed by reduced neutrophil superoxide production and spreading on immobilized anti-ß2 integrin Ab. ATL313 suppressed selectin-triggered activation of Src family kinases (SFKs) and p38 MAPK, chemokine-triggered activation of Ras-related protein 1, and ß2 integrin-triggered activation of SFKs and Vav cytoskeletal regulatory proteins. ATL313 activated protein kinase A and its substrate C-terminal Src kinase, an inhibitor of SFKs. Treating neutrophils with a protein kinase A inhibitor blocked the actions of ATL313. In vivo, ATL313-treated neutrophils rolled faster and arrested much less frequently in postcapillary venules of the murine cremaster muscle after TNF-α challenge. Furthermore, ATL313 markedly suppressed neutrophil migration into the peritoneum challenged with thioglycollate. ATL313 did not affect A2AAR-deficient neutrophils, confirming its specificity. Our findings provide new insights into the anti-inflammatory mechanisms of A2AAR signaling and the potential utility of A2AAR agonists in inflammatory diseases.


Subject(s)
Leukocyte Rolling/immunology , Neutrophils/immunology , Receptor, Adenosine A2A/immunology , Signal Transduction/immunology , Animals , CD18 Antigens/genetics , CD18 Antigens/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Humans , Leukocyte Rolling/drug effects , Mice , Mice, Knockout , Neutrophils/cytology , Piperidines/pharmacology , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/immunology , Receptor, Adenosine A2A/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , src-Family Kinases/genetics , src-Family Kinases/immunology
10.
Trends Immunol ; 36(9): 556-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26282885

ABSTRACT

During infection, a subset of effector T cells seeds the lymphoid and non-lymphoid tissues and gives rise to tissue-resident memory T cells (TRM). Recent findings have provided insight into the molecular and cellular mechanisms underlying tissue instruction of TRM cell homing, as well as the programs involved in their retention and maintenance. We review these findings here, highlighting both common features and distinctions between CD4 TRM and CD8 TRM cells. In this context we examine the role of memory lymphocyte clusters (MLCs), and propose that the MLCs serve as an immediate response center consisting of TRM cells on standby, capable of detecting incoming pathogens and mounting robust local immune responses to contain and limit the spread of infectious agents.


Subject(s)
Immunologic Memory , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cell Survival/immunology , Chemokines/metabolism , Host-Pathogen Interactions/immunology , Humans , Leukocyte Rolling/immunology , Lymphocyte Activation/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Organ Specificity/immunology , Signal Transduction , Transendothelial and Transepithelial Migration/immunology
11.
PLoS One ; 10(8): e0136116, 2015.
Article in English | MEDLINE | ID: mdl-26312490

ABSTRACT

The transforming growth factor beta 1 (TGF-ß1) is a pleiotropic cytokine with multiple roles in development, wound healing, and immune regulation. TGF-ß1-mediated immune dysfunction may lead to pathological conditions, such as inflammation. Chronic inflammatory process is characterized by a continuous release of pro-inflammatory cytokines, and the inhibition or the blockage of these cytokines signaling pathways are considered a target treatment. In this context, despite the high numbers of TGF-ß-targeted pathways, the inducible regulatory T cells (iTreg) to control inflammation seems to be a promising approach. Our aim was to develop novel peptides through phage display (PhD) technology that could mimic TGF-ß1 function with higher potency. Specific mimetic peptides were obtained through a PhD subtraction strategy from whole cell binding using TGF-ß1 recombinant as a competitor during elution step. We have selected a peptide that seems to play an important role on cellular differentiation and modulation of TNF-α and IL-10 cytokines. The synthetic pm26TGF-ß1 peptide tested in PBMC significantly down-modulated TNF-α and up-regulated IL-10 responses, leading to regulatory T cells (Treg) phenotype differentiation. Furthermore, the synthetic peptide was able to decrease leukocytes rolling in BALB/C mice and neutrophils migration during inflammatory process in C57BL/6 mice. These data suggest that this peptide may be useful for the treatment of inflammatory diseases, especially because it displays potent anti-inflammatory properties and do not exhibit neutrophils' chemoattraction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomimetic Materials/pharmacology , Leukocyte Rolling/drug effects , Neutrophils/immunology , Peptides/pharmacology , Transforming Growth Factor beta1/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Biomimetic Materials/chemistry , Female , Humans , Interleukin-10/immunology , Leukocyte Rolling/immunology , Male , Mice , Mice, Inbred BALB C , Neutrophils/pathology , Peptides/chemistry , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta1/chemistry , Tumor Necrosis Factor-alpha/immunology
12.
J Immunol ; 195(3): 1152-61, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101325

ABSTRACT

Neutrophil recruitment to the site of inflammation plays a pivotal role in host defense. However, overwhelming activation and accumulation of neutrophils in the tissue may cause tissue damage and autoimmunity due to the release of cytokines, oxidants, and proteases. Neutrophil adhesion in acute inflammation is initiated by activation of αLß2 (LFA-1), which can be induced by rolling on E-selectin (slowly) or by exposure to the chemokine CXCL1 (rapidly). Despite the clinical importance, cell-intrinsic molecular mechanisms of negative regulation of integrin adhesiveness and neutrophil recruitment are poorly understood. Mice deficient in the tyrosine phosphatase Src homology 2 domain-containing protein tyrosine phosphatase 1 (Shp1) show increased leukocyte adhesion, but the interpretation of these data is limited by the severe global phenotype of these mice. In this study, we used mice with global and myeloid-restricted deletion of Shp1 to study neutrophil arrest, adhesion, crawling, and transendothelial migration in vitro and in vivo. Shp1 deficiency results in increased neutrophil adhesion in vivo; however, neutrophil crawling, transmigration, and chemotaxis were reduced in these mice. Mechanistically, Shp1 binds and controls PIPKIγ activity and, thereby, modulates phosphatidylinositol (4,5)-bisphosphate levels and adhesion. Thus, Shp1 is involved in the deactivation of integrins and regulation of neutrophil recruitment into inflamed tissue.


Subject(s)
Cell Adhesion/immunology , Neutrophil Infiltration/immunology , Phosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor)/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Animals , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Chemokine CXCL1/immunology , E-Selectin/immunology , Enzyme Activation/immunology , HL-60 Cells , Humans , Inflammation/immunology , Leukocyte Rolling/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophils/immunology , Phosphatidylinositols/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
13.
Curr Protein Pept Sci ; 16(1): 77-86, 2015.
Article in English | MEDLINE | ID: mdl-25692849

ABSTRACT

A key event in inflammatory disease is the transendothelial recruitment of leukocytes from the circulation to the site of inflammation. Intense research in the past decades indicates that the polyanionic carbohydrate heparan sulphate (HS) modulates multiple steps in the leukocyte recruitment cascade. Leukocyte recruitment is initiated by endothelial cell activation and presentation of chemokines to rolling leukocytes, which, via integrin activation, results in adhesion and diapedesis through the vessel wall. Heparan sulfate proteoglycans (HSPGs) immobilize the chemokines on the luminal endothelial cells, rendering them more robust against mechanical or hydrodynamic perturbations. During inflammation, endothelial HSPGs serve as ligands to L-selectin on leukocytes, transport chemokines in a basolateral to apical direction across the endothelium, and present chemokines at the luminal surface of the endothelium to circulating cells. HSPGs also promote chemokine oligomerization, which influences chemokine receptor signaling. Furthermore, proteoglycans of the syndecan family are involved in modulating integrin-mediated tight adhesion of leukocytes to the endothelium. Creation of a chemokine gradient by a localized chemokine release influences the speed of leukocyte recruitment from the blood to the tissue by attracting crawling neutrophils to optimal sites for transmigration. The directionality of intraluminal crawling is thought to be influenced by both mechanotactic and haptotactic signals, which are modulated by HS-dependent signaling processes. Finally, diapedesis is influenced by HS regarding transendothelial chemokine gradient formation and integrin- CAM interactions, and further enhanced by heparanase-mediated degradation of the endothelial basement membrane. Overall, the multifunctional role of HS in inflammation marks it as a potential target of glycan-centered therapeutic approaches.


Subject(s)
Chemotaxis, Leukocyte/immunology , Heparitin Sulfate/metabolism , Inflammation/immunology , Inflammation/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Animals , Cell Adhesion/immunology , Humans , Leukocyte Rolling/immunology , Transendothelial and Transepithelial Migration/immunology
14.
J Immunol ; 194(2): 728-38, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25505282

ABSTRACT

Neutrophil recruitment to the site of inflammation plays a pivotal role in host defense. Src family kinases (SFKs) activation is required for integrin and chemokine signaling as well as immune cell function. The receptor-like protein tyrosine phosphatase CD45 positively regulates chemoattractant signaling acting on SFK activity. To further investigate the role of CD45 in neutrophil recruitment and function, we analyzed transgenic mice carrying a single point mutation (CD45E613R), which constitutively activates CD45. By using intravital microscopy experiments, we demonstrated that different steps of the leukocyte recruitment cascade were affected in CD45E613R mutant mice. The rolling velocity of CD45E613R mutant neutrophils was decreased compared with wild-type neutrophils that subsequently resulted in an increased number of adherent cells. The analysis of ß2 integrins LFA-1 and macrophage-1 Ag (Mac-1) showed that in CD45E613R mutant neutrophils LFA-1 adhesiveness was impaired, and avidity was enhanced, whereas Mac-1 adhesiveness was increased. Because of the increased Mac-1 adhesiveness, neutrophil crawling was impaired in CD45E613R mutant compared with wild-type neutrophils. In an Escherichia coli lung infection model, CD45E613R mice displayed a decreased neutrophil recruitment into the alveolar compartment, which resulted in an increased number of CFUs in the lung. Our data demonstrate that the CD45E613R mutation modulates integrin activation and leukocyte recruitment during inflammation.


Subject(s)
Leukocyte Common Antigens , Leukocyte Rolling , Lymphocyte Function-Associated Antigen-1 , Macrophage-1 Antigen , Neutrophils/immunology , Point Mutation , Amino Acid Substitution , Animals , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/immunology , Mice , Mice, Mutant Strains , Neutrophils/pathology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology
15.
J Immunol ; 193(7): 3803-15, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25187658

ABSTRACT

Because ERK5 inhibits endothelial inflammation and dysfunction, activating ERK5 might be a novel approach to protecting vascular endothelial cells (ECs) against various pathological conditions of the blood vessel. We have identified small molecules that protect ECs via ERK5 activation and determined their contribution to preventing cardiac allograft rejection. Using high-throughput screening, we identified certain statins and antimalarial agents including chloroquine, hydroxychloroquine, and quinacrine as strong ERK5 activators. Pitavastatin enhanced ERK5 transcriptional activity and Kruppel-like factor-2 expression in cultured human and bovine ECs, but these effects were abolished by the depletion of ERK5. Chloroquine and hydroxychloroquine upregulated ERK5 kinase activity and inhibited VCAM-1 expression in an ERK5-dependent but MAPK/ERK kinase 5- and Kruppel-like factor 2/4-independent manner. Leukocyte rolling and vascular reactivity were used to evaluate endothelial function in vivo, and we found that EC-specific ERK5 knockout (ERK5-EKO) mice exhibited increased leukocyte rolling and impaired vascular reactivity, which could not be corrected by pitavastatin. The role of endothelial ERK5 in acute cardiac allograft rejection was also examined by heterotopic grafting of the heart obtained from either wild-type or ERK5-EKO mice into allomismatched recipient mice. A robust increase in both inflammatory gene expression and CD45-positive cell infiltration into the graft was observed. These tissue rejection responses were inhibited by pitavastatin in wild-type but not ERK5-EKO hearts. Our study has identified statins and antimalarial drugs as strong ERK5 activators and shown that ERK5 activation is preventive of endothelial inflammation and dysfunction and acute allograft rejection.


Subject(s)
Antimalarials/pharmacology , Endothelium, Vascular/immunology , Graft Rejection/drug therapy , Heart Transplantation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 7/immunology , Quinolines/pharmacology , Transcription, Genetic/drug effects , Allografts , Animals , Cattle , Endothelium, Vascular/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Leukocyte Rolling/drug effects , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 7/genetics , Transcription, Genetic/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
16.
J Exp Med ; 211(9): 1833-46, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25135296

ABSTRACT

The focus of this study is the characterization of human T cell blood-brain barrier migration and corresponding molecular trafficking signatures. We examined peripheral blood and cerebrospinal fluid immune cells from patients under long-term anti-very late antigen-4 (VLA-4)/natalizumab therapy (LTNT) and from CNS specimens. LTNT patients' cerebrospinal fluid T cells exhibited healthy central-/effector-memory ratios, but lacked CD49d and showed enhanced myeloma cell adhesion molecule (MCAM) expression. LTNT led to an increase of PSGL-1 expression on peripheral T cells. Although vascular cell adhesion molecule-1 (VLA-4 receptor) was expressed at all CNS barriers, P-selectin (PSGL-1-receptor) was mainly detected at the choroid plexus. Accordingly, in vitro experiments under physiological flow conditions using primary human endothelial cells and LTNT patients' T cells showed increased PSGL-1-mediated rolling and residual adhesion, even under VLA-4 blockade. Adhesion of MCAM(+)/TH17 cells was not affected by VLA-4 blocking alone, but was abrogated when both VLA-4 and MCAM were inhibited. Consistent with these data, MCAM(+) cells were detected in white matter lesions, and in gray matter of multiple sclerosis patients. Our data indicate that lymphocyte trafficking into the CNS under VLA-4 blockade can occur by using the alternative adhesion molecules, PSGL-1 and MCAM, the latter representing an exclusive pathway for TH17 cells to migrate over the blood-brain barrier.


Subject(s)
Central Nervous System/cytology , Central Nervous System/immunology , Integrin alpha4beta1/antagonists & inhibitors , Membrane Glycoproteins/immunology , T-Lymphocytes/immunology , Th17 Cells/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Blood-Brain Barrier/immunology , CD146 Antigen/immunology , Case-Control Studies , Cell Adhesion/immunology , Cells, Cultured , Humans , Integrin alpha4beta1/immunology , Leukocyte Rolling/immunology , Ligands , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/therapy , Natalizumab , P-Selectin/metabolism , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
17.
Circ Res ; 114(5): 792-805, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24397980

ABSTRACT

RATIONALE: Anti-inflammatory and vascular protective actions of adiponectin are well recognized. However, many fundamental questions remain unanswered. OBJECTIVE: The current study attempted to identify the adiponectin receptor subtype responsible for adiponectin's vascular protective action and investigate the role of ceramidase activation in adiponectin anti-inflammatory signaling. METHODS AND RESULTS: Adiponectin significantly reduced tumor necrosis factor (TNF)α-induced intercellular adhesion molecule-1 expression and attenuated TNFα-induced oxidative/nitrative stress in human umbilical vein endothelial cells. These anti-inflammatory actions were virtually abolished by adiponectin receptor 1 (AdipoR1-), but not AdipoR2-, knockdown (KD). Treatment with adiponectin significantly increased neutral ceramidase (nCDase) activity (3.7-fold; P<0.01). AdipoR1-KD markedly reduced globular adiponectin-induced nCDase activation, whereas AdipoR2-KD only slightly reduced. More importantly, small interfering RNA-mediated nCDase-KD markedly blocked the effect of adiponectin on TNFα-induced intercellular adhesion molecule-1 expression. AMP-activated protein kinase-KD failed to block adiponectin-induced nCDase activation and modestly inhibited adiponectin anti-inflammatory effect. In contrast, in caveolin-1 KD (Cav1-KD) cells, >87% of adiponectin-induced nCDase activation was lost. Whereas adiponectin treatment failed to inhibit TNFα-induced intercellular adhesion molecule-1 expression, treatment with sphingosine-1-phosphate or SEW (sphingosine-1-phosphate receptor agonist) remained effective in Cav1-KD cells. AdipoR1 and Cav1 colocalized and coprecipitated in human umbilical vein endothelial cells. Adiponectin treatment did not affect this interaction. There is weak basal Cav1/nCDase interaction, which significantly increased after adiponectin treatment. Knockout of AdipoR1 or Cav1 abolished the inhibitory effect of adiponectin on leukocyte rolling and adhesion in vivo. CONCLUSIONS: These results demonstrate for the first time that adiponectin inhibits TNFα-induced inflammatory response via Cav1-mediated ceramidase recruitment and activation in an AdipoR1-dependent fashion.


Subject(s)
Adiponectin/metabolism , Caveolin 1/metabolism , Ceramidases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vasculitis/metabolism , Adiponectin/immunology , Caveolin 1/genetics , Caveolin 1/immunology , Ceramidases/genetics , Ceramidases/immunology , Endothelial Cells/immunology , Enzyme Activation/immunology , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Leukocyte Rolling/immunology , RNA, Small Interfering/genetics , Reactive Nitrogen Species/immunology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Receptors, Adiponectin/genetics , Receptors, Adiponectin/immunology , Receptors, Adiponectin/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Vasculitis/immunology
18.
J Immunol ; 192(4): 1862-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24431230

ABSTRACT

Selectins are a family of adhesion receptors designed for efficient leukocyte tethering to the endothelium under shear. As a key property to resist premature bond disruption, selectin adhesiveness is enhanced by tensile forces that promote the conversion of a bent into an extended conformation of the N-terminal lectin and epidermal growth factor-like domains. Conformation-specific Abs have been invaluable in deciphering the activation mechanism of integrins, but similar reagents are not available for selectins. In this study, we show that the anti-human L-selectin mAbs DREG-55 and LAM1-5 but not DREG-56, DREG-200, or LAM1-1 heterotropically modulate adhesion presumably by stabilizing the extended receptor conformation. Force-free affinity assays, flow chamber, and microkinetic studies reveal a ligand-specific modulation of L-selectin affinity by DREG-55 mAb, resulting in a dramatic decrease of rolling velocity under flow. Furthermore, secondary tethering of polymorphonuclear cells was blocked by DREG-200 but significantly boosted by DREG-55 mAb. The results emphasize the need for a new classification for selectin Abs and introduce the new concept of heterotropic modulation of receptor function.


Subject(s)
Antibodies, Monoclonal/immunology , Leukocyte Rolling/immunology , Neutrophils/immunology , Selectins/metabolism , Amino Acid Sequence , Antibodies/immunology , Cell Adhesion/immunology , Cell Line , Humans , Jurkat Cells , Selectins/immunology , Sequence Alignment
19.
Circ Res ; 114(5): 770-9, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24366169

ABSTRACT

RATIONALE: The inflammatory processes that initiate and propagate atherosclerosis remain poorly understood, largely because defining the intravascular behavior of immune cells has been technically challenging. Respiratory and pulsatile movements have hampered in vivo visualization of leukocyte accumulation in athero-prone arteries at resolutions achieved in other tissues. OBJECTIVE: To establish and to validate a method that allows high-resolution imaging of inflammatory leukocytes and platelets within the carotid artery of atherosusceptible mice in vivo. METHODS AND RESULTS: We have devised a procedure to stabilize the mouse carotid artery mechanically without altering blood dynamics, which dramatically enhances temporal and spatial resolutions using high-speed intravital microscopy in multiple channels of fluorescence. By applying this methodology at different stages of disease progression in atherosusceptible mice, we first validated our approach by assessing the recruitment kinetics of various leukocyte subsets and platelets in athero-prone segments of the carotid artery. The high temporal and spatial resolution allowed the dissection of both the dynamic polarization of and the formation of subcellular domains within adhered leukocytes. We further demonstrate that the secondary capture of activated platelets on the plaque is predominantly mediated by neutrophils. Finally, we couple this procedure with triggered 2-photon microscopy to visualize the 3-dimensional movement of leukocytes in intimate contact with the arterial lumen. CONCLUSIONS: The improved imaging of diseased arteries at subcellular resolution presented here should help resolve many outstanding questions in atherosclerosis and other arterial disorders.


Subject(s)
Carotid Artery Diseases/immunology , Carotid Artery Diseases/physiopathology , Microscopy, Fluorescence/methods , Vasculitis/immunology , Vasculitis/physiopathology , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/physiopathology , Blood Platelets/immunology , Carotid Artery Diseases/genetics , Carotid Artery, Common/immunology , Carotid Artery, Common/physiopathology , Female , Green Fluorescent Proteins/genetics , Leukocyte Rolling/immunology , Leukocytes/immunology , Male , Mice , Mice, Knockout , Myeloid Cells/immunology , Neutrophils/immunology , Regional Blood Flow/physiology , Vasculitis/genetics
20.
Blood ; 122(24): 3993-4001, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24106206

ABSTRACT

Beyond its well-established roles in mediating leukocyte rolling, E-selectin is emerging as a multifunctional receptor capable of inducing integrin activation in neutrophils, and of regulating various biological processes in hematopoietic precursors. Although these effects suggest important homeostatic contributions of this selectin in the immune and hematologic systems, the ligands responsible for transducing these effects in different leukocyte lineages are not well defined. We have characterized mice deficient in E-selectin ligand-1 (ESL-1), or in both P-selectin glycoprotein-1 (PSGL-1) and ESL-1, to explore and compare the contributions of these glycoproteins in immune and hematopoietic cell trafficking. In the steady state, ESL-1 deficiency resulted in a moderate myeloid expansion that became more prominent when both glycoproteins were eliminated. During inflammation, PSGL-1 dominated E-selectin binding, rolling, integrin activation, and extravasation of mature neutrophils, but only the combined deficiency in PSGL-1 and ESL-1 completely abrogated leukocyte recruitment. Surprisingly, we find that the levels of ESL-1 were strongly elevated in hematopoietic progenitor cells. These elevations correlated with a prominent function of ESL-1 for E-selectin binding and for migration of hematopoietic progenitor cells into the bone marrow. Our results uncover dominant roles for ESL-1 in the immature compartment, and a functional shift toward PSGL-1 dependence in mature neutrophils.


Subject(s)
Hematopoietic Stem Cells/immunology , Inflammation/immunology , Receptors, Fibroblast Growth Factor/immunology , Sialoglycoproteins/immunology , Animals , Blotting, Western , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Movement/immunology , E-Selectin/metabolism , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Inflammation/genetics , Inflammation/metabolism , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Leukocytes/immunology , Leukocytes/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/metabolism , Protein Binding/immunology , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Sialoglycoproteins/deficiency , Sialoglycoproteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...