Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 143(10): 845-857, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38096370

RESUMO

ABSTRACT: Protease activated receptors (PARs) are cleaved by coagulation proteases and thereby connect hemostasis with innate immune responses. Signaling of the tissue factor (TF) complex with factor VIIa (FVIIa) via PAR2 stimulates extracellular signal-regulated kinase (ERK) activation and cancer cell migration, but functions of cell autonomous TF-FVIIa signaling in immune cells are unknown. Here, we show that myeloid cell expression of FVII but not of FX is crucial for inflammatory cell recruitment to the alveolar space after challenge with the double-stranded viral RNA mimic polyinosinic:polycytidylic acid [Poly(I:C)]. In line with these data, genetically modified mice completely resistant to PAR2 cleavage but not FXa-resistant PAR2-mutant mice are protected from lung inflammation. Poly(I:C)-stimulated migration of monocytes/macrophages is dependent on ERK activation and mitochondrial antiviral signaling (MAVS) but independent of toll-like receptor 3 (TLR3). Monocyte/macrophage-synthesized FVIIa cleaving PAR2 is required for integrin αMß2-dependent migration on fibrinogen but not for integrin ß1-dependent migration on fibronectin. To further dissect the downstream signaling pathway, we generated PAR2S365/T368A-mutant mice deficient in ß-arrestin recruitment and ERK scaffolding. This mutation reduces cytosolic, but not nuclear ERK phosphorylation by Poly(I:C) stimulation, and prevents macrophage migration on fibrinogen but not fibronectin after stimulation with Poly(I:C) or CpG-B, a single-stranded DNA TLR9 agonist. In addition, PAR2S365/T368A-mutant mice display markedly reduced immune cell recruitment to the alveolar space after Poly(I:C) challenge. These results identify TF-FVIIa-PAR2-ß-arrestin-biased signaling as a driver for lung infiltration in response to viral nucleic acids and suggest potential therapeutic interventions specifically targeting TF-VIIa signaling in thrombo-inflammation.


Assuntos
Fator VIIa , Monócitos , Animais , Camundongos , Fator VIIa/metabolismo , Monócitos/metabolismo , Tromboplastina/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Transdução de Sinais/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrinogênio/metabolismo , beta-Arrestinas/metabolismo
2.
Blood ; 143(12): 1167-1180, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38142429

RESUMO

ABSTRACT: Antiphospholipid antibodies (aPL) in primary or secondary antiphospholipid syndrome (APS) are a major cause for acquired thrombophilia, but specific interventions preventing autoimmune aPL development are an unmet clinical need. Although autoimmune aPL cross react with various coagulation regulatory proteins, lipid-reactive aPL, including those derived from patients with COVID-19, recognize the endolysosomal phospholipid lysobisphosphatidic acid presented by the cell surface-expressed endothelial protein C receptor. This specific recognition leads to complement-mediated activation of tissue factor (TF)-dependent proinflammatory signaling and thrombosis. Here, we show that specific inhibition of the TF coagulation initiation complex with nematode anticoagulant protein c2 (NAPc2) prevents the prothrombotic effects of aPL derived from patients with COVID-19 in mice and the aPL-induced proinflammatory and prothrombotic activation of monocytes. The induction of experimental APS is dependent on the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, and NAPc2 suppresses monocyte endosomal reactive oxygen species production requiring the TF cytoplasmic domain and interferon-α secretion from dendritic cells. Latent infection with murine cytomegalovirus causes TF cytoplasmic domain-dependent development of persistent aPL and circulating phospholipid-reactive B1 cells, which is prevented by short-term intervention with NAPc2 during acute viral infection. In addition, treatment of lupus prone MRL-lpr mice with NAPc2, but not with heparin, suppresses dendritic-cell activation in the spleen, aPL production and circulating phospholipid-reactive B1 cells, and attenuates lupus pathology. These data demonstrate a convergent TF-dependent mechanism of aPL development in latent viral infection and autoimmune disease and provide initial evidence that specific targeting of the TF initiation complex has therapeutic benefits beyond currently used clinical anticoagulant strategies.


Assuntos
Síndrome Antifosfolipídica , COVID-19 , Viroses , Humanos , Animais , Camundongos , Anticorpos Antifosfolipídeos , Tromboplastina/metabolismo , Camundongos Endogâmicos MRL lpr , Síndrome Antifosfolipídica/complicações , Fosfolipídeos , Anticoagulantes , COVID-19/complicações , Viroses/complicações
3.
Science ; 371(6534)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33707237

RESUMO

Antiphospholipid antibodies (aPLs) cause severe autoimmune disease characterized by vascular pathologies and pregnancy complications. Here, we identify endosomal lysobisphosphatidic acid (LBPA) presented by the CD1d-like endothelial protein C receptor (EPCR) as a pathogenic cell surface antigen recognized by aPLs for induction of thrombosis and endosomal inflammatory signaling. The engagement of aPLs with EPCR-LBPA expressed on innate immune cells sustains interferon- and toll-like receptor 7-dependent B1a cell expansion and autoantibody production. Specific pharmacological interruption of EPCR-LBPA signaling attenuates major aPL-elicited pathologies and the development of autoimmunity in a mouse model of systemic lupus erythematosus. Thus, aPLs recognize a single cell surface lipid-protein receptor complex to perpetuate a self-amplifying autoimmune signaling loop dependent on the cooperation with the innate immune complement and coagulation pathways.


Assuntos
Apresentação de Antígeno , Autoimunidade , Coagulação Sanguínea/imunologia , Receptor de Proteína C Endotelial/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Lisofosfolipídeos/imunologia , Monoglicerídeos/imunologia , Animais , Anticorpos Antifosfolipídeos/biossíntese , Autoanticorpos/biossíntese , Modelos Animais de Doenças , Perda do Embrião/imunologia , Endossomos/imunologia , Receptor de Proteína C Endotelial/genética , Humanos , Imunidade Inata , Lúpus Eritematoso Sistêmico/sangue , Camundongos , Camundongos Mutantes , Esfingomielina Fosfodiesterase/metabolismo , Trombose/imunologia , Receptor 7 Toll-Like/imunologia
4.
Blood Adv ; 4(22): 5810-5824, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33232477

RESUMO

Deficiencies in many coagulation factors and protease-activated receptors (PARs) affect embryonic development. We describe a defect in definitive erythropoiesis in PAR2-deficient mice. Embryonic PAR2 deficiency increases embryonic death associated with variably severe anemia in comparison with PAR2-expressing embryos. PAR2-deficient fetal livers display reduced macrophage densities, erythroblastic island areas, and messenger RNA expression levels of markers for erythropoiesis and macrophages. Coagulation factor synthesis in the liver coincides with expanding fetal liver hematopoiesis during midgestation, and embryonic factor VII (FVII) deficiency impairs liver macrophage development. Cleavage-insensitive PAR2-mutant mice recapitulate the hematopoiesis defect of PAR2-deficient embryos, and macrophage-expressed PAR2 directly supports erythroblastic island function and the differentiation of red blood cells in the fetal liver. Conditional deletion of PAR2 in macrophages impairs erythropoiesis, as well as increases inflammatory stress, as evidenced by upregulation of interferon-regulated hepcidin antimicrobial peptide. In contrast, postnatal macrophage PAR2 deficiency does not have any effect on steady-state Kupffer cells, bone marrow macrophage numbers, or erythropoiesis, but erythropoiesis in macrophages from PAR2-deficient mice is impaired following hemolysis. These data identify a novel function for macrophage PAR2 signaling in adapting to rapid increases in blood demand during gestational development and postnatal erythropoiesis under stress conditions.


Assuntos
Eritropoese , Fígado , Receptor PAR-2 , Animais , Macrófagos , Camundongos , Camundongos Knockout
5.
Blood ; 132(2): 123-131, 2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-29866813

RESUMO

The hemostatic system plays pivotal roles in injury repair, innate immunity, and adaptation to inflammatory challenges. We review the evidence that these vascular-protective mechanisms have nontraditional roles in hematopoietic stem cell (HSC) maintenance in their physiological bone marrow (BM) niches at steady-state and under stress. Expression of coagulation factors and the extrinsic coagulation initiator tissue factor by osteoblasts, tissue-resident macrophages, and megakaryocytes suggests that endosteal and vascular HSC niches are functionally regulated by extravascular coagulation. The anticoagulant endothelial protein C receptor (EPCR; Procr) is highly expressed by primitive BM HSCs and endothelial cells. EPCR is associated with its major ligand, activated protein C (aPC), in proximity to thrombomodulin-positive blood vessels, enforcing HSC integrin α4 adhesion and chemotherapy resistance in the context of CXCL12-CXCR4 niche retention signals. Protease-activated receptor 1-biased signaling by EPCR-aPC also maintains HSC retention, whereas thrombin signaling activates HSC motility and BM egress. Furthermore, HSC mobilization under stress is enhanced by the fibrinolytic and complement cascades that target HSCs and their BM niches. In addition, coagulation, fibrinolysis, and HSC-derived progeny, including megakaryocytes, synergize to reestablish functional perivascular HSC niches during BM stress. Therapeutic restoration of the anticoagulant pathway has preclinical efficacy in reversing BM failure following radiation injury, but questions remain about how antithrombotic therapy influences extravascular coagulation in HSC maintenance and hematopoiesis.


Assuntos
Coagulação Sanguínea , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Animais , Biomarcadores , Medula Óssea , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Humanos , Transdução de Sinais , Nicho de Células-Tronco , Estresse Fisiológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA