Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Blood ; 143(13): 1293-1309, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38142410

ABSTRACT

ABSTRACT: Although it is caused by a single-nucleotide mutation in the ß-globin gene, sickle cell anemia (SCA) is a systemic disease with complex, incompletely elucidated pathologies. The mononuclear phagocyte system plays critical roles in SCA pathophysiology. However, how heterogeneous populations of hepatic macrophages contribute to SCA remains unclear. Using a combination of single-cell RNA sequencing and spatial transcriptomics via multiplexed error-robust fluorescence in situ hybridization, we identified distinct macrophage populations with diversified origins and biological functions in SCA mouse liver. We previously found that administering the von Willebrand factor (VWF)-cleaving protease ADAMTS13 alleviated vaso-occlusive episode in mice with SCA. Here, we discovered that the ADAMTS13-cleaved VWF was cleared from the circulation by a Clec4f+Marcohigh macrophage subset in a desialylation-dependent manner in the liver. In addition, sickle erythrocytes were phagocytized predominantly by Clec4f+Marcohigh macrophages. Depletion of macrophages not only abolished the protective effect of ADAMTS13 but exacerbated vaso-occlusive episode in mice with SCA. Furthermore, promoting macrophage-mediated VWF clearance reduced vaso-occlusion in SCA mice. Our study demonstrates that hepatic macrophages are important in the pathogenesis of SCA, and efficient clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in SCA mice.


Subject(s)
Anemia, Sickle Cell , Vascular Diseases , Mice , Animals , von Willebrand Factor/genetics , In Situ Hybridization, Fluorescence , Anemia, Sickle Cell/pathology , Macrophages/pathology , ADAMTS13 Protein/genetics
2.
Cells ; 12(8)2023 04 12.
Article in English | MEDLINE | ID: mdl-37190045

ABSTRACT

Integrin LFA-1 plays a critical role in T-cell migration and in the formation of immunological synapses. LFA-1 functions through interacting with its ligands with differing affinities: low, intermediate, and high. Most prior research has studied how LFA-1 in the high-affinity state regulates the trafficking and functions of T cells. LFA-1 is also presented in the intermediate-affinity state on T cells, however, the signaling to activate LFA-1 to the intermediate-affinity state and the role of LFA-1 in this affinity state both remain largely elusive. This review briefly summarizes the activation and roles of LFA-1 with varied ligand-binding affinities in the regulation of T-cell migration and immunological synapse formation.


Subject(s)
Immunological Synapses , Lymphocyte Function-Associated Antigen-1 , Cell Communication , Cell Movement , T-Lymphocytes , Humans , Animals
3.
Proc Natl Acad Sci U S A ; 119(34): e2207592119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969769

ABSTRACT

Vaso-occlusive episode (VOE) is a common and critical complication of sickle cell disease (SCD). Its pathogenesis is incompletely understood. von Willebrand factor (VWF), a multimeric plasma hemostatic protein synthesized and secreted by endothelial cells and platelets, is increased during a VOE. However, whether and how VWF contributes to the pathogenesis of VOE is not fully understood. In this study, we found increased VWF levels during tumor necrosis factor (TNF)-induced VOE in a humanized mouse model of SCD. Deletion of endothelial VWF decreased hemolysis, vascular occlusion, and organ damage caused by TNF-induced VOE in SCD mice. Moreover, administering ADAMTS13, the VWF-cleaving plasma protease, reduced plasma VWF levels, decreased inflammation and vaso-occlusion, and alleviated organ damage during VOE. These data suggest that promoting VWF cleavage via ADAMTS13 may be an effective treatment for reducing hemolysis, inflammation, and vaso-occlusion during VOE.


Subject(s)
Anemia, Sickle Cell , Vascular Diseases , von Willebrand Factor , ADAMTS13 Protein/metabolism , ADAMTS13 Protein/pharmacology , ADAMTS13 Protein/therapeutic use , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Gene Deletion , Hemolysis/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Mice , Vascular Diseases/drug therapy , Vascular Diseases/etiology , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
5.
Blood ; 139(16): 2523-2533, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35157766

ABSTRACT

Microvascular thrombosis in patients with thrombotic thrombocytopenic purpura (TTP) is initiated by GPIbα-mediated platelet binding to von Willebrand factor (VWF). Binding of VWF to GPIbα causes activation of the platelet surface integrin αIIbß3. However, the mechanism of GPIbα-initiated activation of αIIbß3 and its clinical importance for microvascular thrombosis remain elusive. Deletion of platelet C-type lectin-like receptor 2 (CLEC-2) did not prevent VWF binding to platelets but specifically inhibited platelet aggregation induced by VWF binding in mice. Deletion of platelet CLEC-2 also inhibited αIIbß3 activation induced by the binding of VWF to GPIbα. Using a mouse model of TTP, which was created by infusion of anti-mouse ADAMTS13 monoclonal antibodies followed by infusion of VWF, we found that deletion of platelet CLEC-2 decreased pulmonary arterial thrombosis and the severity of thrombocytopenia. Importantly, prophylactic oral administration of aspirin, an inhibitor of platelet activation, and therapeutic treatment of the TTP mice with eptifibatide, an integrin αIIbß3 antagonist, reduced pulmonary arterial thrombosis in the TTP mouse model. Our observations demonstrate that GPIbα-mediated activation of integrin αIIbß3 plays an important role in the formation of thrombosis in TTP. These observations suggest that prevention of platelet activation with aspirin may reduce the risk for thrombosis in patients with TTP.


Subject(s)
Hypertension, Pulmonary , Purpura, Thrombotic Thrombocytopenic , Thrombosis , Aspirin , Blood Platelets/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Purpura, Thrombotic Thrombocytopenic/metabolism , Thrombosis/etiology , von Willebrand Factor/metabolism
6.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34291736

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a pandemic. Severe disease is associated with dysfunction of multiple organs, but some infected cells do not express ACE2, the canonical entry receptor for SARS-CoV-2. Here, we report that the C-type lectin receptor L-SIGN interacted in a Ca2+-dependent manner with high-mannose-type N-glycans on the SARS-CoV-2 spike protein. We found that L-SIGN was highly expressed on human liver sinusoidal endothelial cells (LSECs) and lymph node lymphatic endothelial cells but not on blood endothelial cells. Using high-resolution confocal microscopy imaging, we detected SARS-CoV-2 viral proteins within the LSECs from liver autopsy samples from patients with COVID-19. We found that both pseudo-typed virus enveloped with SARS-CoV-2 spike protein and authentic SARS-CoV-2 virus infected L-SIGN-expressing cells relative to control cells. Moreover, blocking L-SIGN function reduced CoV-2-type infection. These results indicate that L-SIGN is a receptor for SARS-CoV-2 infection. LSECs are major sources of the clotting factors vWF and factor VIII (FVIII). LSECs from liver autopsy samples from patients with COVID-19 expressed substantially higher levels of vWF and FVIII than LSECs from uninfected liver samples. Our data demonstrate that L-SIGN is an endothelial cell receptor for SARS-CoV-2 that may contribute to COVID-19-associated coagulopathy.


Subject(s)
COVID-19 , Capillaries , Cell Adhesion Molecules/metabolism , Endothelial Cells , Lectins, C-Type/metabolism , Liver/blood supply , Lymphatic Vessels , Receptors, Cell Surface/metabolism , SARS-CoV-2/physiology , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , Capillaries/metabolism , Capillaries/pathology , Capillaries/virology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/virology , Gene Expression Profiling/methods , Humans , Liver/pathology , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Lymphatic Vessels/virology , Spike Glycoprotein, Coronavirus , Virus Internalization
7.
Cell Death Differ ; 28(11): 3009-3021, 2021 11.
Article in English | MEDLINE | ID: mdl-33993195

ABSTRACT

The liver has recently been identified as a major organ for destruction of desialylated platelets. However, the underlying mechanism remains unclear. Kupffer cells, which are professional phagocytic cells in the liver, comprise the largest population of resident tissue macrophages in the body. Kupffer cells express a C-type lectin receptor, CLEC4F, that recognizes desialylated glycans with an unclear in vivo role in mediating platelet destruction. In this study, we generated a CLEC4F-deficient mouse model (Clec4f-/-) and found that CLEC4F was specifically expressed by Kupffer cells. Using the Clec4f-/- mice and a newly generated platelet-specific reporter mouse line, we revealed a critical role for CLEC4F on Kupffer cells in mediating destruction of desialylated platelets in the liver in vivo. Platelet clearance experiments and ultrastructural analysis revealed that desialylated platelets were phagocytized predominantly by Kupffer cells in a CLEC4F-dependent manner in mice. Collectively, these findings identify CLEC4F as a Kupffer cell receptor important for the destruction of desialylated platelets induced by bacteria-derived neuraminidases, which provide new insights into the pathogenesis of thrombocytopenia in disease conditions such as sepsis.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Blood Platelets/metabolism , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Animals , Disease Models, Animal , Mice
8.
Blood ; 137(20): 2756-2769, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33619517

ABSTRACT

During early embryonic development in mammals, including humans and mice, megakaryocytes (Mks) first originate from primitive hematopoiesis in the yolk sac. These embryonic Mks (eMks) circulate in the vasculature with unclear function. Herein, we report that podoplanin (PDPN), the ligand of C-type lectin-like receptor (CLEC-2) on Mks/platelets, is temporarily expressed in neural tissue during midgestation in mice. Loss of PDPN or CLEC-2 resulted in aneurysms and spontaneous hemorrhage, specifically in the lower diencephalon during midgestation. Surprisingly, more eMks/platelets had enhanced granule release and localized to the lower diencephalon in mutant mouse embryos than in wild-type littermates before hemorrhage. We found that PDPN counteracted the collagen-1-induced secretion of angiopoietin-1 from fetal Mks, which coincided with enhanced TIE-2 activation in aneurysm-like sprouts of PDPN-deficient embryos. Blocking platelet activation prevented the PDPN-deficient embryo from developing vascular defects. Our data reveal a new role for PDPN in regulating eMk function during midgestation.


Subject(s)
Brain/blood supply , Intracranial Aneurysm/etiology , Megakaryocytes/pathology , Membrane Glycoproteins/deficiency , Aneurysm, Ruptured/embryology , Aneurysm, Ruptured/etiology , Angiopoietin-1/metabolism , Animals , Brain/embryology , Cells, Cultured , Cerebral Hemorrhage/embryology , Cerebral Hemorrhage/etiology , Collagen/pharmacology , Diencephalon/blood supply , Diencephalon/embryology , Gene Expression Regulation, Developmental , Gestational Age , Intracranial Aneurysm/embryology , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lectins, C-Type/physiology , Megakaryocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Platelet Activation , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptor, TIE-2/metabolism
9.
Science ; 370(6515): 467-472, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33093110

ABSTRACT

Colon mucus segregates the intestinal microbiota from host tissues, but how it organizes to function throughout the colon is unclear. In mice, we found that colon mucus consists of two distinct O-glycosylated entities of Muc2: a major form produced by the proximal colon, which encapsulates the fecal material including the microbiota, and a minor form derived from the distal colon, which adheres to the major form. The microbiota directs its own encapsulation by inducing Muc2 production from proximal colon goblet cells. In turn, O-glycans on proximal colon-derived Muc2 modulate the structure and function of the microbiota as well as transcription in the colon mucosa. Our work shows how proximal colon control of mucin production is an important element in the regulation of host-microbiota symbiosis.


Subject(s)
Colon/metabolism , Colon/microbiology , Gastrointestinal Microbiome , Mucin-2/metabolism , Mucus/metabolism , Animals , Feces/microbiology , Glycosylation , Mice , Mice, Knockout , Mucin-2/genetics , Transcription, Genetic
10.
Blood Adv ; 4(15): 3559-3571, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32761234

ABSTRACT

Ezrin/radixin/moesin (ERM) proteins are adaptors that link the actin cytoskeleton to the cytoplasmic domains of membrane proteins. Leukocytes express mostly moesin with lower levels of ezrin but no radixin. When leukocytes are activated, ERMs are postulated to redistribute membrane proteins from microvilli into uropods during polarization and to transduce signals that influence adhesion and other responses. However, these functions have not been tested in leukocytes lacking all ERMs. We used knockout (KO) mice with neutrophils lacking ezrin, moesin, or both proteins (double knockout [DKO]) to probe how ERMs modulate cell shape, adhesion, and signaling in vitro and in vivo. Surprisingly, chemokine-stimulated DKO neutrophils still polarized and redistributed ERM-binding proteins such as PSGL-1 and CD44 to the uropods. Selectin binding to PSGL-1 on moesin KO or DKO neutrophils activated kinases that enable integrin-dependent slow rolling but not those that generate neutrophil extracellular traps. Flowing neutrophils of all genotypes rolled normally on selectins and, upon chemokine stimulation, arrested on integrin ligands. However, moesin KO and DKO neutrophils exhibited defective integrin outside-in signaling and reduced adhesion strength. In vivo, DKO neutrophils displayed normal directional crawling toward a chemotactic gradient, but premature detachment markedly reduced migration from venules into inflamed tissues. Our results demonstrate that stimulated neutrophils do not require ERMs to polarize or to move membrane proteins into uropods. They also reveal an unexpected contribution of moesin to integrin outside-in signaling and adhesion strengthening.


Subject(s)
Membrane Proteins , Neutrophils , Animals , Cytoskeletal Proteins , Membrane Proteins/genetics , Mice , Microfilament Proteins
11.
JCI Insight ; 5(13)2020 07 09.
Article in English | MEDLINE | ID: mdl-32641582

ABSTRACT

Multiple organ failure in sepsis is a progressive failure of several interdependent organ systems. Liver dysfunction occurs early during sepsis and is directly associated with patient death; however, the underlying mechanism of liver dysfunction is unclear. Platelet transfusion benefits patients with sepsis, and inhibition of complement activation protects liver function in septic animals. Herein, we explored the potential link between platelets, complement activation, and liver dysfunction in sepsis. We found that deletion of platelet C-type lectin-like receptor 2 (CLEC-2) exacerbated liver dysfunction in early sepsis. Platelet CLEC-2-deficient mice exhibited higher complement activation, more severe complement attack in the liver, and lower plasma levels of complement inhibitors at early time points after E. coli infection. Circulating monocytes expressed the CLEC-2 ligand podoplanin in early sepsis, and podoplanin binding induced release of complement inhibitors from platelets. Injection of complement inhibitors released from platelets reduced complement attack and attenuated liver dysfunction in septic mice. These findings indicate a new function of platelets in the regulation of complement activation during sepsis.


Subject(s)
Complement Inactivating Agents/pharmacology , Liver/drug effects , Membrane Glycoproteins/metabolism , Monocytes/metabolism , Sepsis/complications , Animals , Blood Platelets/metabolism , Complement Inactivating Agents/metabolism , Liver/metabolism , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/genetics , Mice , Monocytes/drug effects , Platelet Activation/drug effects , Platelet Activation/physiology , Sepsis/chemically induced , Up-Regulation
12.
J Immunol ; 204(1): 37-48, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31757864

ABSTRACT

During inflammation, both neutrophils and effector T cells use selectins to roll and integrins to arrest in postcapillary venules. In both cell types, chemokines can transduce signals that convert integrin αLß2 to a high-affinity conformation, which interacts with ICAM-1 to mediate arrest. In neutrophils, selectins also trigger an immunoreceptor-like signaling cascade that converts integrin αLß2 to an intermediate-affinity conformation, which interacts with ICAM-1 to slow rolling. It is not known whether selectins induce similar signaling events in T cells. Ag engagement causes phosphorylation of ITAMs on the TCR; these motifs recruit kinases and adaptors that lead to the activation of αLß2. We found that mouse Th1 cells rolling on P- or E-selectin triggered signals that promoted αLß2-dependent slow rolling on ICAM-1 in vitro and in vivo. The selectin signaling cascade resembled that used by the TCR, except that unexpectedly, Th1 cells employed the ITAM-bearing protein DAP12, which was not known to be expressed in these cells. Importantly, outside-in signaling through ligand-occupied αLß2 also required DAP12. Cooperative selectin and chemokine signaling in Th1 cells promoted αLß2-dependent slow rolling and arrest in vitro and in vivo and migration into Ag-challenged tissues in vivo. Our findings reveal an important function for DAP12 in Th1 cells and a new mechanism to recruit effector T cells to sites of inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Th1 Cells/immunology , Animals , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Proc Natl Acad Sci U S A ; 114(31): 8360-8365, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716912

ABSTRACT

Most platelet membrane proteins are modified by mucin-type core 1-derived glycans (O-glycans). However, the biological importance of O-glycans in platelet clearance is unclear. Here, we generated mice with a hematopoietic cell-specific loss of O-glycans (HC C1galt1-/- ). These mice lack O-glycans on platelets and exhibit reduced peripheral platelet numbers. Platelets from HC C1galt1-/- mice show reduced levels of α-2,3-linked sialic acids and increased accumulation in the liver relative to wild-type platelets. The preferential accumulation of HC C1galt1-/- platelets in the liver was reduced in mice lacking the hepatic asialoglycoprotein receptor [Ashwell-Morell receptor (AMR)]. However, we found that Kupffer cells are the primary cells phagocytosing HC C1galt1-/- platelets in the liver. Our results demonstrate that hepatic AMR promotes preferential adherence to and phagocytosis of desialylated and/or HC C1galt1-/- platelets by the Kupffer cell through its C-type lectin receptor CLEC4F. These findings provide insights into an essential role for core 1 O-glycosylation of platelets in their clearance in the liver.


Subject(s)
Blood Platelets/metabolism , Galactosyltransferases/genetics , Kupffer Cells/metabolism , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Animals , Asialoglycoprotein Receptor/metabolism , Hepatocytes/metabolism , Homeostasis/physiology , Lectins, C-Type/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Thrombocytopenia/pathology
14.
Blood ; 130(2): 181-191, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28515093

ABSTRACT

Leukocyte adhesion to P-selectin on activated platelets and endothelial cells induces shedding of the P-selectin ectodomain into the circulation. Plasma soluble P-selectin (sP-selectin) is elevated threefold to fourfold in patients with cardiovascular disease. Circulating sP-selectin is thought to trigger signaling in leukocytes that directly contributes to inflammation and thrombosis. However, sP-selectin likely circulates as a monomer, and in vitro studies suggest that sP-selectin must dimerize to induce signaling in leukocytes. To address this discrepancy, we expressed the entire ectodomain of mouse P-selectin as a monomer (sP-selectin) or as a disulfide-linked dimer fused to the Fc portion of mouse immunoglobulin G (sP-selectin-Fc). Dimeric sP-selectin-Fc, but not monomeric sP-selectin, triggered integrin-dependent adhesion of mouse leukocytes in vitro. Antibody-induced oligomerization of sP-selectin or sP-selectin-Fc was required to trigger formation of neutrophil extracellular traps. Injecting sP-selectin-Fc, but not sP-selectin, into mice augmented integrin-dependent adhesion of neutrophils in venules, generated tissue factor-bearing microparticles, shortened plasma-clotting times, and increased thrombus frequency in the inferior vena cava. Furthermore, transgenic mice that overexpressed monomeric sP-selectin did not exhibit increased inflammation or thrombosis. We conclude that elevated plasma sP-selectin is a consequence rather than a cause of cardiovascular disease.


Subject(s)
Extracellular Traps/immunology , Neutrophils/immunology , P-Selectin/blood , Thrombosis/genetics , Vena Cava, Inferior/immunology , Animals , Antibodies/pharmacology , CD18 Antigens/genetics , CD18 Antigens/immunology , CHO Cells , Cell Adhesion/drug effects , Cricetulus , Disulfides/chemistry , Extracellular Traps/drug effects , Gene Expression Regulation , Immunoglobulin Fc Fragments/blood , Immunoglobulin Fc Fragments/genetics , Inflammation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/pathology , P-Selectin/chemistry , P-Selectin/genetics , P-Selectin/immunology , Protein Domains , Protein Multimerization , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Thromboplastin/genetics , Thromboplastin/immunology , Thrombosis/immunology , Thrombosis/pathology , Vena Cava, Inferior/pathology
15.
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
16.
Proc Natl Acad Sci U S A ; 112(28): 8661-6, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26124096

ABSTRACT

Palmitoylated cysteines typically target transmembrane proteins to domains enriched in cholesterol and sphingolipids (lipid rafts). P-selectin glycoprotein ligand-1 (PSGL-1), CD43, and CD44 are O-glycosylated proteins on leukocytes that associate with lipid rafts. During inflammation, they transduce signals by engaging selectins as leukocytes roll in venules, and they move to the raft-enriched uropods of polarized cells upon chemokine stimulation. It is not known how these glycoproteins associate with lipid rafts or whether this association is required for signaling or for translocation to uropods. Here, we found that loss of core 1-derived O-glycans in murine C1galt1(-/-) neutrophils blocked raft targeting of PSGL-1, CD43, and CD44, but not of other glycosylated proteins, as measured by resistance to solubilization in nonionic detergent and by copatching with a raft-resident sphingolipid on intact cells. Neuraminidase removal of sialic acids from wild-type neutrophils also blocked raft targeting. C1galt1(-/-) neutrophils or neuraminidase-treated neutrophils failed to activate tyrosine kinases when plated on immobilized anti-PSGL-1 or anti-CD44 F(ab')2. Furthermore, C1galt1(-/-) neutrophils incubated with anti-PSGL-1 F(ab')2 did not generate microparticles. In marked contrast, PSGL-1, CD43, and CD44 moved normally to the uropods of chemokine-stimulated C1galt1(-/-) neutrophils. These data define a role for core 1-derived O-glycans and terminal sialic acids in targeting glycoprotein ligands for selectins to lipid rafts of leukocytes. Preassociation of these glycoproteins with rafts is required for signaling but not for movement to uropods.


Subject(s)
Leukocytes/metabolism , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Polysaccharides/metabolism , Animals , Hyaluronan Receptors/metabolism , Leukosialin/metabolism , Ligands , Mice
17.
J Exp Med ; 212(8): 1267-81, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26169939

ABSTRACT

Neutrophil recruitment, mediated by ß2 integrins, combats pyogenic infections but also plays a key role in ischemia-reperfusion injury and other inflammatory disorders. Talin induces allosteric rearrangements in integrins that increase affinity for ligands (activation). Talin also links integrins to actin and other proteins that enable formation of adhesions. Structural studies have identified a talin1 mutant (L325R) that perturbs activation without impairing talin's capacity to link integrins to actin and other proteins. Here, we found that mice engineered to express only talin1(L325R) in myeloid cells were protected from renal ischemia-reperfusion injury. Dissection of neutrophil function in vitro and in vivo revealed that talin1(L325R) neutrophils had markedly impaired chemokine-induced, ß2 integrin-mediated arrest, spreading, and migration. Surprisingly, talin1(L325R) neutrophils exhibited normal selectin-induced, ß2 integrin-mediated slow rolling, in sharp contrast to the defective slow rolling of neutrophils lacking talin1 or expressing a talin1 mutant (W359A) that blocks talin interaction with integrins. These studies reveal the importance of talin-mediated activation of integrins for renal ischemia-reperfusion injury. They further show that neutrophil arrest requires talin recruitment to and activation of integrins. However, although neutrophil slow rolling requires talin recruitment to integrins, talin-mediated integrin activation is dispensable.


Subject(s)
CD18 Antigens/metabolism , Kidney Diseases/prevention & control , Leukocyte Rolling/physiology , Neutrophils/physiology , Reperfusion Injury/prevention & control , Talin/metabolism , Animals , Blotting, Western , Cell Movement/genetics , DNA Primers/genetics , Immunoprecipitation , Kidney Diseases/genetics , Mice , Mice, Transgenic , Mutation, Missense/genetics , Neutrophils/metabolism , Reperfusion Injury/genetics , Talin/genetics
18.
Blood ; 122(23): 3832-42, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24081661

ABSTRACT

Neutrophils emigrate from venules to sites of infection or injury in response to chemotactic gradients. How these gradients form is not well understood. Some IL-6 family cytokines stimulate endothelial cells to express adhesion molecules and chemokines that recruit leukocytes. Receptors for these cytokines share the signaling subunit gp130. We studied knockout mice lacking gp130 in endothelial cells. Unexpectedly, gp130-deficient endothelial cells constitutively expressed more CXCL1 in vivo and in vitro, and even more upon stimulation with tumor necrosis factor-α. Mobilization of this increased CXCL1 from intracellular stores to the venular surface triggered ß2 integrin-dependent arrest of neutrophils rolling on selectins but impaired intraluminal crawling and transendothelial migration. Superfusing CXCL1 over venules promoted neutrophil migration only after intravenously injecting mAb to CXCL1 to diminish its intravascular function or heparinase to release CXCL1 from endothelial proteoglycans. Remarkably, mice lacking gp130 in endothelial cells had impaired histamine-induced venular permeability, which was restored by injecting anti-P-selectin mAb to prevent neutrophil rolling and arrest. Thus, excessive CXCL1 expression in gp130-deficient endothelial cells augments neutrophil adhesion but hinders migration, most likely by disrupting chemotactic gradients. Our data define a role for endothelial cell gp130 in regulating integrin-dependent adhesion and de-adhesion of neutrophils during inflammation.


Subject(s)
Chemokine CXCL1/metabolism , Cytokine Receptor gp130/deficiency , Endothelial Cells/physiology , Neutrophils/physiology , Animals , Capillary Permeability/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Chemokine CXCL1/genetics , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/physiology , Inflammation/physiopathology , Leukocyte Rolling/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/physiology , Up-Regulation , Venules/physiology
19.
J Biol Chem ; 287(23): 19585-98, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22511754

ABSTRACT

In inflamed venules, neutrophils roll on P- or E-selectin, engage P-selectin glycoprotein ligand-1 (PSGL-1), and signal extension of integrin α(L)ß(2) in a low affinity state to slow rolling on intercellular adhesion molecule-1 (ICAM-1). Cytoskeleton-dependent receptor clustering often triggers signaling, and it has been hypothesized that the cytoplasmic domain links PSGL-1 to the cytoskeleton. Chemokines cause rolling neutrophils to fully activate α(L)ß(2), leading to arrest on ICAM-1. Cytoskeletal anchorage of α(L)ß(2) has been linked to chemokine-triggered extension and force-regulated conversion to the high affinity state. We asked whether PSGL-1 must interact with the cytoskeleton to initiate signaling and whether α(L)ß(2) must interact with the cytoskeleton to extend. Fluorescence recovery after photobleaching of transfected cells documented cytoskeletal restraint of PSGL-1. The lateral mobility of PSGL-1 similarly increased by depolymerizing actin filaments with latrunculin B or by mutating the cytoplasmic tail to impair binding to the cytoskeleton. Converting dimeric PSGL-1 to a monomer by replacing its transmembrane domain did not alter its mobility. By transducing retroviruses expressing WT or mutant PSGL-1 into bone marrow-derived macrophages from PSGL-1-deficient mice, we show that PSGL-1 required neither dimerization nor cytoskeletal anchorage to signal ß(2) integrin-dependent slow rolling on P-selectin and ICAM-1. Depolymerizing actin filaments or decreasing actomyosin tension in neutrophils did not impair PSGL-1- or chemokine-mediated integrin extension. Unlike chemokines, PSGL-1 did not signal cytoskeleton-dependent swing out of the ß(2)-hybrid domain associated with the high affinity state. The cytoskeletal independence of PSGL-1-initiated, α(L)ß(2)-mediated slow rolling differs markedly from the cytoskeletal dependence of chemokine-initiated, α(L)ß(2)-mediated arrest.


Subject(s)
Leukocyte Rolling/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Glycoproteins/metabolism , Neutrophils/metabolism , Signal Transduction/physiology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actomyosin/genetics , Actomyosin/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CHO Cells , Chemokines/genetics , Chemokines/metabolism , Cricetinae , Cricetulus , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neutrophils/cytology , Protein Multimerization/drug effects , Protein Multimerization/physiology , Protein Structure, Tertiary , Signal Transduction/drug effects , Thiazolidines/pharmacology
20.
Blood ; 118(15): 4015-23, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21860019

ABSTRACT

Trousseau syndrome is classically defined as migratory, heparin-sensitive but warfarin-resistant microthrombi in patients with occult, mucinous adenocarcinomas. Injecting carcinoma mucins into mice generates platelet-rich microthrombi dependent on P- and L-selectin but not thrombin. Heparin prevents mucin binding to P- and L-selectin and mucin-induced microthrombi. This model of Trousseau syndrome explains resistance to warfarin, which inhibits fluid-phase coagulation but not selectins. Here we found that carcinoma mucins do not generate microthrombi in mice lacking P-selectin glycoprotein ligand-1 (PSGL-1), the leukocyte ligand for P- and L-selectin. Furthermore, mucins did not activate platelets in blood from PSGL-1-deficient mice. Mucins induced microthrombi in radiation chimeras lacking endothelial P-selectin but not in chimeras lacking platelet P-selectin. Mucins caused leukocytes to release cathepsin G, but only if platelets were present. Mucins failed to generate microthrombi in cathepsin G-deficient mice. Mucins did not activate platelets in blood from mice lacking cathepsin G or protease-activated receptor-4 (PAR4), indicating that cathepsin G activates platelets through PAR4. Using knockout mice and blocking antibodies, we found that mucin-triggered cathepsin G release requires L-selectin and PSGL-1 on neutrophils, P-selectin on platelets, and Src family kinases in both cell types. Thus, carcinoma mucins promote thrombosis through adhesion-dependent, bidirectional signaling in neutrophils and platelets.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Blood Platelets/metabolism , Mucins/metabolism , Neoplasm Proteins/metabolism , Neutrophil Activation , Neutrophils/metabolism , Platelet Activation , Thrombosis/metabolism , Adenocarcinoma, Mucinous/complications , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Animals , Antibodies, Neoplasm/pharmacology , Antibodies, Neutralizing/pharmacology , Blood Platelets/pathology , Cathepsin G/genetics , Cathepsin G/metabolism , Cell Line, Tumor , Colonic Neoplasms , Disease Models, Animal , Humans , L-Selectin/genetics , L-Selectin/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Mucins/antagonists & inhibitors , Mucins/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neutrophils/pathology , P-Selectin/genetics , P-Selectin/metabolism , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Syndrome , Thrombosis/etiology , Thrombosis/genetics , Thrombosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...