Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Blood ; 140(9): 1020-1037, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35737916

RESUMO

Acute lung injury, referred to as the acute chest syndrome, is a major cause of morbidity and mortality in patients with sickle cell disease (SCD), which often occurs in the setting of a vaso-occlusive painful crisis. P-selectin antibody therapy reduces hospitalization of patients with SCD by ∼50%, suggesting that an unknown P-selectin-independent mechanism promotes remaining vaso-occlusive events. In patients with SCD, intraerythrocytic polymerization of mutant hemoglobin promotes ischemia-reperfusion injury and hemolysis, which leads to the development of sterile inflammation. Using intravital microscopy in transgenic, humanized mice with SCD and in vitro studies with blood from patients with SCD, we reveal for the first time that the sterile inflammatory milieu in SCD promotes caspase-4/11-dependent activation of neutrophil-gasdermin D (GSDMD), which triggers P-selectin-independent shedding of neutrophil extracellular traps (NETs) in the liver. Remarkably, these NETs travel intravascularly from liver to lung, where they promote neutrophil-platelet aggregation and the development of acute lung injury. This study introduces a novel paradigm that liver-to-lung embolic translocation of NETs promotes pulmonary vascular vaso-occlusion and identifies a new GSDMD-mediated, P-selectin-independent mechanism of lung injury in SCD.


Assuntos
Lesão Pulmonar Aguda , Anemia Falciforme , Armadilhas Extracelulares , Proteínas de Ligação a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Traumatismo por Reperfusão , Lesão Pulmonar Aguda/etiologia , Animais , Fígado , Pulmão/irrigação sanguínea , Camundongos , Camundongos Transgênicos , Selectina-P , Proteínas de Ligação a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Traumatismo por Reperfusão/complicações
2.
Haematologica ; 2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-37941440

RESUMO

Sickle cell disease (SCD) is a monogenic disorder that affects 100,000 African Americans and millions of people worldwide. Intra-erythrocytic polymerization of sickle hemoglobin (HbS) promotes erythrocyte sickling, impaired rheology, ischemia and hemolysis, leading to the development of progressive liver injury in SCD. Liver resident macrophages and monocytes are known to enable the clearance of HbS, however, the role of liver sinusoidal endothelial cells (LSECs) in HbS clearance and liver injury in SCD remains unknown. Using real-time intravital (in vivo) imaging in the mice liver as well as flow cytometric analysis and confocal imaging of primary human LSECs, we show for the first time that liver injury in SCD is associated with accumulation of HbS and iron in the LSECs, leading to LSEC senescence. Hb uptake by LSECs was mediated by micropinocytosis. Hepatic monocytes were observed to attenuate LSECsenescence by accelerating HbS clearance in the liver of SCD mice, however, this protection was impaired in P-selectin-deficient SCD mice secondary to reduced monocyte recruitment in the liver. These findings are the first to suggest that LSECs contribute to HbS clearance and HbS induced LSEC-senescence promotes progressive liver injury in SCD mice. Our results provide a novel insight into the pathogenesis of hemolysis induced chronic liver injury in SCD caused by LSEC senescence. Identifying the regulators of LSEC mediated HbS clearance may lead to new therapies to prevent the progression of liver injury in SCD.

3.
Blood ; 135(20): 1783-1787, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31977004

RESUMO

Vaso-occlusive crisis (VOC) is the primary cause of morbidity and hospitalization in sickle cell disease (SCD); however, only 4 therapies (hydroxyurea, l-glutamine, crizanlizumab, and voxeletor) are currently approved in SCD. These agents limit the duration, severity, and frequency of crises. Activation of coagulation is a hallmark of SCD. Studies in animal models of SCD have shown that coagulation contributes to the chronic inflammation and end-organ damage associated with the disease; however, it is unknown whether coagulation directly contributes to the microvascular stasis that causes VOC. Herein, we demonstrate that inhibition of tissue factor (TF) and the downstream coagulation proteases factor Xa and thrombin significantly attenuates heme-induced microvascular stasis in mouse models of VOC. Pharmacologic inhibition of the principal thrombin receptor, protease activated receptor-1 (PAR-1), as well as deficiency of PAR-1 in all nonhematopoietic cells, also reduces stasis in sickle mice. PAR-1 deficiency was associated with reduced endothelial von Willebrand factor expression, which has been shown to mediate microvascular stasis. In addition, TF inhibition reduces lung vaso-occlusion in sickle mice mediated by arteriolar neutrophil-platelet microemboli. In sum, these results suggest that prophylactic anticoagulation might attenuate the incidence of VOC.


Assuntos
Anemia Falciforme/metabolismo , Transtornos da Coagulação Sanguínea/etiologia , Receptor PAR-1/metabolismo , Trombina/metabolismo , Anemia Falciforme/complicações , Anemia Falciforme/genética , Anemia Falciforme/patologia , Animais , Transtornos da Coagulação Sanguínea/genética , Transtornos da Coagulação Sanguínea/metabolismo , Plaquetas/metabolismo , Constrição Patológica/genética , Constrição Patológica/metabolismo , Modelos Animais de Doenças , Feminino , Hemoglobina Falciforme/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microvasos/metabolismo , Microvasos/patologia , Receptor PAR-1/genética , Doenças Vasculares/etiologia , Doenças Vasculares/metabolismo
4.
Circ Res ; 127(5): 677-692, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32493166

RESUMO

RATIONALE: Unproven theories abound regarding the long-range uptake and endocrine activity of extracellular blood-borne microRNAs into tissue. In pulmonary hypertension (PH), microRNA-210 (miR-210) in pulmonary endothelial cells promotes disease, but its activity as an extracellular molecule is incompletely defined. OBJECTIVE: We investigated whether chronic and endogenous endocrine delivery of extracellular miR-210 to pulmonary vascular endothelial cells promotes PH. METHODS AND RESULTS: Using miR-210 replete (wild-type [WT]) and knockout mice, we tracked blood-borne miR-210 using bone marrow transplantation and parabiosis (conjoining of circulatory systems). With bone marrow transplantation, circulating miR-210 was derived predominantly from bone marrow. Via parabiosis during chronic hypoxia to induce miR-210 production and PH, miR-210 was undetectable in knockout-knockout mice pairs. However, in plasma and lung endothelium, but not smooth muscle or adventitia, miR-210 was observed in knockout mice of WT-knockout pairs. This was accompanied by downregulation of miR-210 targets ISCU (iron-sulfur assembly proteins)1/2 and COX10 (cytochrome c oxidase assembly protein-10), indicating endothelial import of functional miR-210. Via hemodynamic and histological indices, knockout-knockout pairs were protected from PH, whereas knockout mice in WT-knockout pairs developed PH. In particular, pulmonary vascular engraftment of miR-210-positive interstitial lung macrophages was observed in knockout mice of WT-knockout pairs. To address whether engrafted miR-210-positive myeloid or lymphoid cells contribute to paracrine miR-210 delivery, we studied miR-210 knockout mice parabiosed with miR-210 WT; Cx3cr1 knockout mice (deficient in myeloid recruitment) or miR-210 WT; Rag1 knockout mice (deficient in lymphocytes). In both pairs, miR-210 knockout mice still displayed miR-210 delivery and PH, thus demonstrating a pathogenic endocrine delivery of extracellular miR-210. CONCLUSIONS: Endogenous blood-borne transport of miR-210 into pulmonary vascular endothelial cells promotes PH, offering fundamental insight into the systemic physiology of microRNA activity. These results also describe a platform for RNA-mediated crosstalk in PH, providing an impetus for developing blood-based miR-210 technologies for diagnosis and therapy in this disease.


Assuntos
Endotélio Vascular/metabolismo , Hipertensão Pulmonar/metabolismo , Pulmão/irrigação sanguínea , MicroRNAs/metabolismo , Animais , Transplante de Medula Óssea , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/fisiopatologia , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/fisiopatologia , Hipóxia/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/sangue , MicroRNAs/genética , Parabiose , Transdução de Sinais
5.
Hepatology ; 72(6): 2165-2181, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32190913

RESUMO

BACKGROUND AND AIMS: Hepatic crisis is an emergent complication affecting patients with sickle cell disease (SCD); however, the molecular mechanism of sickle cell hepatobiliary injury remains poorly understood. Using the knock-in humanized mouse model of SCD and SCD patient blood, we sought to mechanistically characterize SCD-associated hepato-pathophysiology applying our recently developed quantitative liver intravital imaging, RNA sequence analysis, and biochemical approaches. APPROACH AND RESULTS: SCD mice manifested sinusoidal ischemia, progressive hepatomegaly, liver injury, hyperbilirubinemia, and increased ductular reaction under basal conditions. Nuclear factor kappa B (NF-κB) activation in the liver of SCD mice inhibited farnesoid X receptor (FXR) signaling and its downstream targets, leading to loss of canalicular bile transport and altered bile acid pool. Intravital imaging revealed impaired bile secretion into the bile canaliculi, which was secondary to loss of canalicular bile transport and bile acid metabolism, leading to intrahepatic bile accumulation in SCD mouse liver. Blocking NF-κB activation rescued FXR signaling and partially ameliorated liver injury and sinusoidal ischemia in SCD mice. CONCLUSIONS: These findings identify that NF-κB/FXR-dependent impaired bile secretion promotes intrahepatic bile accumulation, which contributes to hepatobiliary injury of SCD. Improved understanding of these processes could potentially benefit the development of therapies to treat sickle cell hepatic crisis.


Assuntos
Anemia Falciforme/complicações , Bile/metabolismo , Colestase/etiologia , Insuficiência Hepática/etiologia , Fígado/patologia , Adolescente , Adulto , Anemia Falciforme/sangue , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Animais , Ductos Biliares Intra-Hepáticos/diagnóstico por imagem , Ductos Biliares Intra-Hepáticos/patologia , Colestase/patologia , Colestase/prevenção & controle , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Hemoglobina Falciforme/genética , Insuficiência Hepática/patologia , Insuficiência Hepática/prevenção & controle , Humanos , Microscopia Intravital , Fígado/diagnóstico por imagem , Masculino , Camundongos , Pessoa de Meia-Idade , NF-kappa B/antagonistas & inibidores , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem
6.
Am J Respir Crit Care Med ; 201(1): 33-46, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31498653

RESUMO

Rationale: Intraerythrocytic polymerization of Hb S promotes hemolysis and vasoocclusive events in the microvasculature of patients with sickle cell disease (SCD). Although platelet-neutrophil aggregate-dependent vasoocclusion is known to occur in the lung and contribute to acute chest syndrome, the etiological mechanisms that trigger acute chest syndrome are largely unknown.Objectives: To identify the innate immune mechanism that promotes platelet-neutrophil aggregate-dependent lung vasoocclusion and injury in SCD.Methods:In vivo imaging of the lung in transgenic humanized SCD mice and in vitro imaging of SCD patient blood flowing through a microfluidic system was performed. SCD mice were systemically challenged with nanogram quantities of LPS to trigger lung vasoocclusion.Measurements and Main Results: Platelet-inflammasome activation led to generation of IL-1ß and caspase-1-carrying platelet extracellular vesicles (EVs) that bind to neutrophils and promote platelet-neutrophil aggregation in lung arterioles of SCD mice in vivo and SCD human blood in microfluidics in vitro. The inflammasome activation, platelet EV generation, and platelet-neutrophil aggregation were enhanced by the presence of LPS at a nanogram dose in SCD but not control human blood. Inhibition of the inflammasome effector caspase-1 or IL-1ß pathway attenuated platelet EV generation, prevented platelet-neutrophil aggregation, and restored microvascular blood flow in lung arterioles of SCD mice in vivo and SCD human blood in microfluidics in vitro.Conclusions: These results are the first to identify that platelet-inflammasome-dependent shedding of IL-1ß and caspase-1-carrying platelet EVs promote lung vasoocclusion in SCD. The current findings also highlight the therapeutic potential of targeting the platelet-inflammasome-dependent innate immune pathway to prevent acute chest syndrome.


Assuntos
Anemia Falciforme/complicações , Anemia Falciforme/imunologia , Vesículas Extracelulares/imunologia , Inflamassomos/imunologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/fisiopatologia , Agregação Plaquetária/imunologia , Síndrome Torácica Aguda/etiologia , Síndrome Torácica Aguda/fisiopatologia , Anemia Falciforme/fisiopatologia , Animais , Humanos , Camundongos , Camundongos Transgênicos , Modelos Animais , Neutrófilos/imunologia
8.
JCI Insight ; 9(2)2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38060312

RESUMO

Cigarette smoking is associated with a higher risk of ICU admissions among patients with flu. However, the etiological mechanism by which cigarette smoke (CS) exacerbates flu remains poorly understood. Here, we show that a mild dose of influenza A virus promotes a severe lung injury in mice preexposed to CS but not room air for 4 weeks. Real-time intravital (in vivo) lung imaging revealed that the development of acute severe respiratory dysfunction in CS- and flu-exposed mice was associated with the accumulation of platelet-rich neutrophil-platelet aggregates (NPAs) in the lung microcirculation within 2 days following flu infection. These platelet-rich NPAs formed in situ and grew larger over time to occlude the lung microvasculature, leading to the development of pulmonary ischemia followed by the infiltration of NPAs and vascular leakage into the alveolar air space. These findings suggest, for the first time to our knowledge, that an acute onset of platelet-driven thrombo-inflammatory response in the lung contributes to the development of CS-induced severe flu.


Assuntos
Fumar Cigarros , Neutrófilos , Humanos , Animais , Camundongos , Fumar Cigarros/efeitos adversos , Pulmão , Plaquetas , Produtos do Tabaco
9.
Blood ; 118(11): 3182-5, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21791417

RESUMO

In a previous study, we demonstrated unique secretory dynamics of tissue plasminogen activator (tPA) in which tPA was retained on the cell surface in a heavy chain-dependent manner after exocytosis from secretory granules in vascular endothelial cells. Here, we examined how retained tPA expresses its enzymatic activity. Retained tPA effectively increased the lysine binding site-dependent binding of plasminogen on the cell surface and pericellular area; this was abolished by inhibition of enzymatic activity of either tPA or plasmin, which suggests that de novo generation of carboxyl-terminal lysine as a consequence of degradation of surface/pericellular proteins by plasmin is essential. Retained tPA initiated zonal clot lysis of a fibrin network that had been formed on vascular endothelial cells, which was preceded by the binding of plasminogen to the lysis front. Our results provide evidence that secreted and retained tPA is essential for maintaining both high fibrinolytic activity and effective clot lysis on the vascular endothelial cell surface.


Assuntos
Células Endoteliais/metabolismo , Fibrinólise , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tecidual/fisiologia , Antígenos de Superfície/metabolismo , Células Cultivadas , Eficiência , Células Endoteliais/fisiologia , Fibrinólise/genética , Fibrinólise/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Transporte Proteico/fisiologia , Ativador de Plasminogênio Tecidual/genética , Transfecção
10.
Arterioscler Thromb Vasc Biol ; 32(9): 2149-57, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22772756

RESUMO

OBJECTIVE: We compared the antithrombotic effects in vivo of 2 chemically different carbon monoxide-releasing molecules (CORM-A1 and CORM-3) on arterial and venous thrombus formation and on hemostatic parameters such as platelet activation, coagulation, and fibrinolysis. The hypotensive response to CORMs and their effects on whole blood gas analysis and blood cell count were also examined. METHODS AND RESULTS: CORM-A1 (10-30 µmol/kg, i.v.), in a dose-dependent fashion, significantly decreased weight of electrically induced thrombus in rats, whereas CORM-3 inhibited thrombosis only at the highest dose used (30 µmol/kg). CORM-A1 showed a direct and stronger inhibition of platelet aggregation than CORM-3 in healthy rats, both in vitro and in vivo. The antiaggregatory effect of CORM-A1, but not CORM-3, correlated positively with weight of the thrombus. Concentration of active plasminogen activator inhibitor-1 in plasma also decreased in response to CORM-A1, but not to CORM-3. Neither CORM-A1 nor CORM-3 had an effect on plasma concentration of active tissue plasminogen activator. CORM-3, but not CORM-A1, decreased the concentration of fibrinogen, fibrin generation, and prolonged prothrombin time. Similarly, laser-induced venous thrombosis observed intravitally via confocal system in green fluorescent protein mice was significantly decreased by CORMs. Although both CORM-A1 and CORM-3 (30 µmol/kg) decreased platelets accumulation in thrombus, only CORM-A1 (3-30 µmol/kg) inhibited platelet activation to phosphatidylserine on their surface. CONCLUSIONS: CORM-3 and CORM-A1 inhibited thrombosis in vivo, however CORM-A1, which slowly releases carbon monoxide, and displayed a relatively weak hypotensive effect had a more pronounced antithrombotic effect associated with a stronger inhibition of platelet aggregation associated with a decrease in active plasminogen activator inhibitor-1 concentration. In contrast, the fast CO releaser CORM-3 that displayed a more pronounced hypotensive effect inhibited thrombosis primarily through a decrease in fibrin generation, but had no direct influence on platelet aggregation and fibrynolysis.


Assuntos
Arteriopatias Oclusivas/prevenção & controle , Boranos/farmacologia , Monóxido de Carbono/metabolismo , Carbonatos/farmacologia , Fibrinolíticos/farmacologia , Compostos Organometálicos/farmacologia , Trombose/prevenção & controle , Trombose Venosa/prevenção & controle , Água/química , Animais , Arteriopatias Oclusivas/sangue , Arteriopatias Oclusivas/etiologia , Arteriopatias Oclusivas/fisiopatologia , Coagulação Sanguínea/efeitos dos fármacos , Gasometria , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Boranos/administração & dosagem , Boranos/química , Boranos/metabolismo , Carbonatos/administração & dosagem , Carbonatos/química , Carbonatos/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibrina/metabolismo , Fibrinogênio/metabolismo , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/administração & dosagem , Fibrinolíticos/química , Fibrinolíticos/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/química , Compostos Organometálicos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/sangue , Agregação Plaquetária/efeitos dos fármacos , Tempo de Protrombina , Ratos , Ratos Wistar , Solubilidade , Trombose/sangue , Trombose/etiologia , Trombose/fisiopatologia , Fatores de Tempo , Trombose Venosa/sangue , Trombose Venosa/etiologia , Trombose Venosa/fisiopatologia
11.
Cells ; 11(3)2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-35159179

RESUMO

Cigarette smoke (CS) is the most common risk factor for chronic obstructive pulmonary disease (COPD). The present study aimed to elucidate whether mtDNA is released upon CS exposure and is detected in the plasma of former smokers affected by COPD as a possible consequence of airway damage. We measured cell-free mtDNA (cf-mtDNA) and nuclear DNA (cf-nDNA) in COPD patient plasma and mouse serum with CS-induced emphysema. The plasma of patients with COPD and serum of mice with CS-induced emphysema showed increased cf-mtDNA levels. In cell culture, exposure to a sublethal dose of CSE decreased mitochondrial membrane potential, increased oxidative stress, dysregulated mitochondrial dynamics, and triggered mtDNA release in extracellular vesicles (EVs). Mitochondrial DNA release into EVs occurred concomitantly with increased expression of markers that associate with DNA damage responses, including DNase III, DNA-sensing receptors (cGAS and NLRP3), proinflammatory cytokines (IL-1ß, IL-6, IL-8, IL-18, and CXCL2), and markers of senescence (p16 and p21); the majority of the responses are also triggered by cytosolic DNA delivery in vitro. Exposure to a lethal CSE dose preferentially induced mtDNA and nDNA release in the cell debris. Collectively, the results of this study associate markers of mitochondrial stress, inflammation, and senescence with mtDNA release induced by CSE exposure. Because high cf-mtDNA is detected in the plasma of COPD patients and serum of mice with emphysema, our findings support the future study of cf-mtDNA as a marker of mitochondrial stress in response to CS exposure and COPD pathology.


Assuntos
Fumar Cigarros , Enfisema , Doença Pulmonar Obstrutiva Crônica , Enfisema Pulmonar , Animais , Fumar Cigarros/efeitos adversos , DNA Mitocondrial , Humanos , Camundongos , Doença Pulmonar Obstrutiva Crônica/metabolismo , Enfisema Pulmonar/genética , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patologia , Nicotiana/genética
12.
Blood Adv ; 6(12): 3729-3734, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35427414

RESUMO

Hemophilia A is an inherited bleeding disorder caused by defective or deficient coagulation factor VIII (FVIII) activity. Until recently, the only treatment for prevention of bleeding involved IV administration of FVIII. Gene therapy with adeno-associated vectors (AAVs) has shown some efficacy in patients with hemophilia A. However, limitations persist due to AAV-induced cellular stress, immunogenicity, and reduced durability of gene expression. Herein, we examined the efficacy of liver-directed gene transfer in FVIII knock-out mice by AAV8-GFP. Surprisingly, compared with control mice, FVIII knockout (F8TKO) mice showed significant delay in AAV8-GFP transfer in the liver. We found that the delay in liver-directed gene transfer in F8TKO mice was associated with absence of liver sinusoidal endothelial cell (LSEC) fenestration, which led to aberrant expression of several sinusoidal endothelial proteins, causing increased capillarization and decreased permeability of LSECs. This is the first study to link impaired liver-directed gene transfer to liver-endothelium maladaptive structural changes associated with FVIII deficiency in mice.


Assuntos
Hemofilia A , Animais , Endotélio , Terapia Genética , Vetores Genéticos/genética , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/terapia , Humanos , Fígado/metabolismo , Camundongos , Camundongos Knockout
13.
Sci Adv ; 8(26): eabm9138, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35767602

RESUMO

The up-regulation of kynurenine metabolism induces immunomodulatory responses via incompletely understood mechanisms. We report that increases in cellular and systemic kynurenine levels yield the electrophilic derivative kynurenine-carboxyketoalkene (Kyn-CKA), as evidenced by the accumulation of thiol conjugates and saturated metabolites. Kyn-CKA induces NFE2 like bZIP transcription factor 2- and aryl hydrocarbon receptor-regulated genes and inhibits nuclear factor κB- and NLR family pyrin domain containing 3-dependent proinflammatory signaling. Sickle cell disease (SCD) is a hereditary hemolytic condition characterized by basal inflammation and recurrent vaso-occlusive crises. Both transgenic SCD mice and patients with SCD exhibit increased kynurenine and Kyn-CKA metabolite levels. Plasma hemin and kynurenine concentrations are positively correlated, indicating that Kyn-CKA synthesis in SCD is up-regulated during pathogenic vascular stress. Administration of Kyn-CKA abrogated pulmonary microvasculature occlusion in SCD mice, an important factor in lung injury development. These findings demonstrate that the up-regulation of kynurenine synthesis and its metabolism to Kyn-CKA is an adaptive response that attenuates inflammation and protects tissues.

14.
Pflugers Arch ; 461(6): 623-33, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21494805

RESUMO

To elucidate how unusually large von Willebrand factor (UL-VWF) multimers facilitate thrombus formation, their behavior was analyzed together with that of platelets in living mice deficient in the gene encoding the protease that cleaves UL-VWF, a disintegrin-like and metalloprotease with thrombospondin type 1 motif 13 (ADAMTS13-/-). By crossing ADAMTS13-/- mice with green fluorescent protein-expressing transgenic mice (GFP mice), GFP-ADAMTS13-/- mice were obtained. The dynamics of GFP-expressing platelets were monitored employing intravital confocal fluorescent microscopy. Administration of a vasopressin derivative, DDAVP, a secretagogue of VWF increased the number of platelets adhered to vascular endothelial cells (VECs) on mesentery at sites recognized by an anti-VWF antibody. Some of these platelets were interconnected and aligned as beads on a string. They reached their maximum length at 5 min and were longer in GFP-ADAMTS13-/- mice than in GFP mice (5.3 ± 4.3, N = 6 vs 2.9 ± 2.1 µm, N = 4) (mean±SE). Focal injury of VECs by topical application of FeCl(3) developed longer (25, 3-50 vs 10, 2-25 µm, P < 0.01) (mean, 10th-90th percentile) and more stable (1.3, 0.3-6.3 vs 0.3, 0.2-1.3 s, P < 0.01) connected platelets in GFP-ADAMTS13-/- mice than in GFP mice. This study revealed that ADAMTS13 cleaves platelet-bound UL-VWF multimers, both during their secretion from VECs and after their adherence to injured vascular walls in veins. UL-VWF multimers either being secreted from VECs or circulating in plasma of ADAMTS13-/- mice appeared to facilitate the accumulation of longer and more stable VWF strings with more associated platelets on injured vascular walls.


Assuntos
Plaquetas/metabolismo , Endotélio Vascular/metabolismo , Fator de von Willebrand/metabolismo , Proteína ADAMTS13 , Animais , Desamino Arginina Vasopressina/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos da radiação , Lasers , Metaloendopeptidases/deficiência , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Adesividade Plaquetária , Multimerização Proteica , Trombose/etiologia
15.
Curr Protoc Cytom ; 95(1): e80, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33226733

RESUMO

Live imaging is critical to determining the dynamics and spatial interactions of cells within the tissue environment. In the lung, this has proven to be difficult due to the motion brought about by ventilation and cardiac contractions. A previous version of this Current Protocols in Cytometry article reported protocols for imaging ex vivo live lung slices and the intact mouse lung. Here, we update those protocols by adding new methodologies, new approaches for quantitative image analysis, and new areas of potential application. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Live imaging of lung slices Support Protocol 1: Staining lung sections with fluorescent antibodies Basic Protocol 2: Live imaging in the mouse lung Support Protocol 2: Intratracheal instillations Support Protocol 3: Intravascular instillations Support Protocol 4: Monitoring vital signs of the mouse during live lung imaging Support Protocol 5: Antibodies Support Protocol 6: Fluorescent reporter mice Basic Protocol 3: Quantification of neutrophil-platelet aggregation in pulmonary vasculature Basic Protocol 4: Quantification of platelet-dependent pulmonary thrombosis Basic Protocol 5: Quantification of pulmonary vascular permeability.


Assuntos
Diagnóstico por Imagem , Pulmão/diagnóstico por imagem , Animais , Diagnóstico por Imagem/métodos , Camundongos , Coloração e Rotulagem
16.
JCI Insight ; 5(14)2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32544100

RESUMO

Patients with hereditary or acquired hemolytic anemias have a high risk of developing in situ thrombosis of the pulmonary vasculature. While pulmonary thrombosis is a major morbidity associated with hemolytic disorders, the etiological mechanism underlying hemolysis-induced pulmonary thrombosis remains largely unknown. Here, we use intravital lung microscopy in mice to assess the pathogenesis of pulmonary thrombosis following deionized water-induced acute intravascular hemolysis. Acute hemolysis triggered the development of αIIbß3-dependent platelet-rich thrombi in precapillary pulmonary arterioles, which led to the transient impairment of pulmonary blood flow. The hemolysis-induced pulmonary thrombosis was phenocopied with intravascular ADP- but not thrombin-triggered pulmonary thrombosis. Consistent with a mechanism involving ADP release from hemolyzing erythrocytes, the inhibition of platelet P2Y12 purinergic receptor signaling attenuated pulmonary thrombosis and rescued blood flow in the pulmonary arterioles of mice following intravascular hemolysis. These findings are the first in vivo studies to our knowledge to suggest that acute intravascular hemolysis promotes ADP-dependent platelet activation, leading to thrombosis in the precapillary pulmonary arterioles, and that thrombin generation most likely does not play a significant role in the pathogenesis of acute hemolysis-triggered pulmonary thrombosis.


Assuntos
Anemia Hemolítica/genética , Hemólise/genética , Artéria Pulmonar/metabolismo , Receptores Purinérgicos P2Y12/genética , Trombose/genética , Difosfato de Adenosina/metabolismo , Anemia Hemolítica/metabolismo , Anemia Hemolítica/patologia , Animais , Arteríolas/metabolismo , Arteríolas/patologia , Coagulação Sanguínea/genética , Plaquetas/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Artéria Pulmonar/patologia , Trombina/genética , Trombina/metabolismo , Trombose/metabolismo , Trombose/patologia
17.
J Thromb Haemost ; 17(10): 1733-1745, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31294514

RESUMO

BACKGROUND: Traumatic injury can lead to dysregulation of the normal clotting system, resulting in hemorrhagic and thrombotic complications. Platelet activation is robust following traumatic injury and one process of platelet activation is to release of extracellular vesicles (PEV) that carry heterogenous cargo loads and surface ligands. OBJECTIVES: We sought to investigate and characterize the release and function of PEVs generated following traumatic injury. METHODS: PEV content and quantity in circulation following trauma in humans and mice was measured using flow cytometry, size exclusion chromatography, and nanoparticle tracking analysis. PEVs were isolated from circulation and the effects on thrombin generation, bleeding time, hemorrhage control, and thrombus formation were determined. Finally, the effect of hydroxychloroquine (HCQ) on PEV release and thrombosis were examined. RESULTS: Human and murine trauma results in a significant release of PEVs into circulation compared with healthy controls. These PEVs result in abundant thrombin generation, increased platelet aggregation, decreased bleeding times, and decreased hemorrhage in uncontrolled bleeding. Conversely, PEVs contributed to enhanced venous thrombus formation and were recruited to the developing thrombus site. Interestingly, HCQ treatment resulted in decreased platelet aggregation, decreased PEV release, and reduced deep vein thrombosis burden in mice. CONCLUSIONS: These data demonstrate that trauma results in significant release of PEVs which are both pro-hemostatic and pro-thrombotic. The effects of PEVs can be mitigated by treatment with HCQ, suggesting the potential use as a form of deep vein thrombosis prophylaxis.


Assuntos
Plaquetas/metabolismo , Vesículas Extracelulares/metabolismo , Hemostasia , Traumatismo Múltiplo/complicações , Trombose Venosa/etiologia , Adulto , Idoso , Animais , Plaquetas/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fibrinolíticos/farmacologia , Hemostasia/efeitos dos fármacos , Humanos , Hidroxicloroquina/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Traumatismo Múltiplo/sangue , Traumatismo Múltiplo/tratamento farmacológico , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Transdução de Sinais , Trombina/metabolismo , Fatores de Tempo , Trombose Venosa/sangue , Trombose Venosa/prevenção & controle
18.
Mil Med Res ; 5(1): 24, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30056803

RESUMO

BACKGROUND: Acute lung injury (ALI) is a major component of multiple organ dysfunction syndrome (MODS) following pulmonary and systemic infection. Alveolar macrophages (AMϕ) are at the center of ALI pathogenesis. Emerging evidence has shown that cell-cell interactions in the lungs play an important regulatory role in the development of acute lung inflammation. However, the underneath mechanisms remain poorly addressed. In this study, we explore a novel function of lung epithelial cells (LEPCs) in regulating the release of exosomes from AMϕ following LPS stimulation. METHODS: For the in vivo experiments, C57BL/6 wildtype (WT) mice were treated with lipopolysaccharide (LPS) (2 mg/kg B.W.) in 0.2 ml of saline via intratracheal aerosol administration. Bronchoalveolar lavage fluid was collected at 0-24 h after LPS treatment, and exosomes derived from AMϕ were measured. For the in vitro studies, LEPCs and bone marrow-derived Mϕ (BMDM) were isolated from WT or TLR4-/- mice and were then cocultured in the Transwell™ system. After coculture for 0-24 h, the BMDM and supernatant were harvested for the measurement of exosomes and cytokines. RESULTS: We demonstrate that LPS induces macrophages (Mϕ) to release exosomes, which are then internalized by neighboring Mϕ to promote TNF-α expression. The secreted interleukin (IL)-25 from LEPCs downregulates Rab27a and Rab27b expression in Mϕ, resulting in suppressed exosome release and thereby attenuating exosome-induced TNF-α expression and secretion. CONCLUSION: These findings reveal a previously unidentified crosstalk pathway between LEPCs and Mϕ that negatively regulates the inflammatory responses of Mϕ to LPS. Modulating IL-25 signaling and targeting exosome release may present a new therapeutic strategy for the treatment of ALI.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Exossomos/metabolismo , Interleucina-17/metabolismo , Lipopolissacarídeos , Macrófagos Alveolares/metabolismo , Lesão Pulmonar Aguda/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/citologia , Células Epiteliais/metabolismo , Inflamação/metabolismo , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais , Fator de Necrose Tumoral alfa/efeitos adversos , Proteínas rab27 de Ligação ao GTP/metabolismo
19.
Thromb Haemost ; 117(4): 682-690, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28150854

RESUMO

Using intravital confocal microscopy, we observed previously that the process of platelet phosphatidylserine (PS) exposure, fibrin formation and lysine binding site-dependent plasminogen (plg) accumulation took place only in the centre of thrombi, not at their periphery. These findings prompted us to analyse the spatiotemporal regulatory mechanisms underlying coagulation and fibrinolysis. We analysed the fibrin network formation and the subsequent lysis in an in vitro experiment using diluted platelet-rich plasma supplemented with fluorescently labelled coagulation and fibrinolytic factors, using confocal laser scanning microscopy. The structure of the fibrin network formed by supplemented tissue factor was uneven and denser at the sites of coagulation initiation regions (CIRs) on PS-exposed platelets. When tissue-type plasminogen activator (tPA; 7.5 nM) was supplemented, labelled plg (50 nM) as well as tPA accumulated at CIRs, from where fibrinolysis started and gradually expanded to the peripheries. The lysis time at CIRs and their peripheries (50 µm from the CIR) were 27.9 ± 6.6 and 44.4 ± 9.7 minutes (mean ± SD, n=50 from five independent experiments) after the addition of tissue factor, respectively. Recombinant human soluble thrombomodulin (TMα; 2.0 nM) attenuated the CIR-dependent plg accumulation and strongly delayed fibrinolysis at CIRs. A carboxypeptidase inhibitor dose-dependently enhanced the CIR-dependent fibrinolysis initiation, and at 20 µM it completely abrogated the TMα-induced delay of fibrinolysis. Our findings are the first to directly present crosstalk between coagulation and fibrinolysis, which takes place on activated platelets' surface and is further controlled by thrombin-activatable fibrinolysis inhibitor (TAFI).


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Carboxipeptidase B2/farmacologia , Fibrinólise/efeitos dos fármacos , Microscopia Intravital/métodos , Microscopia Confocal/métodos , Ativação Plaquetária/efeitos dos fármacos , Plasma Rico em Plaquetas/diagnóstico por imagem , Plaquetas/enzimologia , Carboxipeptidase B2/antagonistas & inibidores , Relação Dose-Resposta a Droga , Fibrina/metabolismo , Humanos , Fosfatidilserinas/metabolismo , Plasma Rico em Plaquetas/citologia , Plasma Rico em Plaquetas/enzimologia , Inibidores de Proteases/farmacologia , Trombomodulina/metabolismo , Fatores de Tempo
20.
FEBS Open Bio ; 6(5): 469-76, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27419052

RESUMO

We reported that tissue-type plasminogen activator (tPA) secreted from vascular endothelial cells (VECs) is retained on the cell surface and effectively evokes both plasminogen activation and fibrin clot dissolution (fibrinolysis) on VECs. Here, to evaluate possibly different behaviors of variants of tPA, we quantitatively assessed these two events separately using green fluorescent protein (GFP)-conjugated tPA in cultured human VECs. The amount of secreted wild-type tPA-GFP correlated well with both the activities of plasminogen activation (r = 0.66) and fibrinolysis (r = -0.93). A variant of tPA-GFP, with a lower affinity to the surface of VECs but higher affinity to fibrin, showed higher fibrinolysis and lower plasminogen activation activity compared to the wild-type.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA