Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Biomedicines ; 11(10)2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37893058

RESUMO

Facioscapulohumeral dystrophy (FSHD) is a muscle disease caused by inappropriate expression of the double homeobox 4 (DUX4) gene in skeletal muscle, and its downstream activation of pro-apoptotic transcriptional programs. Inhibitors of DUX4 expression have the potential to treat FSHD. Apabetalone is a clinical-stage bromodomain and extra-terminal (BET) inhibitor, selective for the second bromodomain on BET proteins. Using primary human skeletal muscle cells from FSHD type 1 patients, we evaluated apabetalone for its ability to counter DUX4's deleterious effects and compared it with the pan-BET inhibitor JQ1, and the p38 MAPK inhibitor-and DUX4 transcriptional repressor-losmapimod. We applied RNA-sequencing and bioinformatic analysis to detect treatment-associated impacts on the transcriptome of these cells. Apabetalone inhibited the expression of DUX4 downstream markers, reversing hallmarks of FSHD gene expression in differentiated muscle cells. JQ1, but not apabetalone, was found to induce apoptosis. While both BET inhibitors modestly impacted differentiation marker expression, they did not affect myotube fusion. Losmapimod also reduced expression of DUX4 target genes but differed in its impact on FSHD-associated pathways. These findings demonstrate that apabetalone inhibits DUX4 target gene expression and reverses transcriptional programs that contribute to FSHD pathology, making this drug a promising candidate therapeutic for FSHD.

2.
Biomedicines ; 11(6)2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37371758

RESUMO

Epigenetic mechanisms are implicated in transcriptional programs driving chronic kidney disease (CKD). Apabetalone is an orally available inhibitor of bromodomain and extraterminal (BET) proteins, which are epigenetic readers that modulate gene expression. In the phase 3 BETonMACE trial, apabetalone reduced risk of major adverse cardiac events (MACE) by 50% in the CKD subpopulation, indicating favorable effects along the kidney-heart axis. Activation of human renal mesangial cells (HRMCs) to a contractile phenotype that overproduces extracellular matrix (ECM) and inflammatory cytokines, and promotes calcification, frequently accompanies CKD to drive pathology. Here, we show apabetalone downregulated HRMC activation with TGF-ß1 stimulation by suppressing TGF-ß1-induced α-smooth muscle actin (α-SMA) expression, α-SMA assembly into stress fibers, enhanced contraction, collagen overproduction, and expression of key drivers of fibrosis, inflammation, or calcification including thrombospondin, fibronectin, periostin, SPARC, interleukin 6, and alkaline phosphatase. Lipopolysaccharide-stimulated expression of inflammatory genes IL6, IL1B, and PTGS2 was also suppressed. Transcriptomics confirmed apabetalone affected gene sets of ECM remodeling and integrins. Clinical translation of in vitro results was indicated in CKD patients where a single dose of apabetalone reduced plasma levels of key pro-fibrotic and inflammatory markers, and indicated inhibition of TGF-ß1 signaling. While plasma proteins cannot be traced to the kidney alone, anti-fibrotic and anti-inflammatory effects of apabetalone identified in this study are consistent with the observed decrease in cardiovascular risk in CKD patients.

3.
Thromb Res ; 231: 182-194, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36307228

RESUMO

Platelets are anucleated cells derived from megakaryocytes that are primarily responsible for hemostasis. However, in recent years, these cytoplasts have become increasingly recognized as immune cells, able to detect, interact with, and kill pathogens. As platelets are involved in both immunity and coagulation, they have a central role in immunothrombosis, a physiological process in which immune cells induce the formation of microthrombi to both prevent the spread of pathogens, and to help facilitate clearance. In this review, we will highlight the role of platelets as key players in the inflammatory and innate immune response against bacterial and viral infection, including direct and indirect interactions with pathogens and other immune cells.


Assuntos
Plaquetas , Inflamação , Humanos , Imunidade Inata , Coagulação Sanguínea , Hemostasia
4.
Front Cell Infect Microbiol ; 12: 897019, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35811684

RESUMO

Hemolytic uremic syndrome (HUS) is the most common cause of acute renal failure in the pediatric population. The etiology of HUS is linked to Gram-negative, Shiga toxin (Stx)-producing enterohemorrhagic bacterial infections. While the effect of Stx is focused on endothelial damage of renal glomerulus, cytokines induced by Stx or bacterial lipopolysaccharide (LPS) and polymorphonuclear cells (PMNs) are involved in the development of the disease. PMN release neutrophil extracellular traps (NETs) to eliminate pathogens, although NETs favor platelets (Plts) adhesion/thrombus formation and can cause tissue damage within blood vessels. Since thrombus formation and occlusion of vessels are characteristic of HUS, PMN-Plts interaction in the context of Stx may promote netosis and contribute to the endothelial damage observed in HUS. The aim of this study was to determine the relevance of netosis induced by Stx in the context of LPS-sensitized Plts on endothelial damage. We observed that Stx2 induced a marked enhancement of netosis promoted by Plts after LPS stimulation. Several factors seemed to promote this phenomenon. Stx2 itself increased the expression of its receptor on Plts, increasing toxin binding. Stx2 also increased LPS binding to Plts. Moreover, Stx2 amplified LPS induced P-selectin expression on Plts and mixed PMN-Plts aggregates formation, which led to activation of PMN enhancing dramatically NETs formation. Finally, experiments revealed that endothelial cell damage mediated by PMN in the context of Plts treated with LPS and Stx2 was decreased when NETs were disrupted or when mixed aggregate formation was impeded using an anti-P-selectin antibody. Using a murine model of HUS, systemic endothelial damage/dysfunction was decreased when NETs were disrupted, or when Plts were depleted, indicating that the promotion of netosis by Plts in the context of LPS and Stx2 plays a fundamental role in endothelial toxicity. These results provide insights for the first time into the pivotal role of Plts as enhancers of endothelial damage through NETs promotion in the context of Stx and LPS. Consequently, therapies designed to reduce either the formation of PMN-Plts aggregates or NETs formation could lessen the consequences of endothelial damage in HUS.


Assuntos
Armadilhas Extracelulares , Síndrome Hemolítico-Urêmica , Trombose , Animais , Criança , Células Endoteliais/metabolismo , Armadilhas Extracelulares/metabolismo , Humanos , Lipopolissacarídeos/toxicidade , Camundongos , Toxina Shiga , Trombose/complicações
5.
Front Immunol ; 12: 772859, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858432

RESUMO

The influenza A virus (IAV) causes a respiratory tract infection with approximately 10% of the population infected by the virus each year. Severe IAV infection is characterized by excessive inflammation and tissue pathology in the lungs. Platelet and neutrophil recruitment to the lung are involved in the pathogenesis of IAV, but the specific mechanisms involved have not been clarified. Using confocal intravital microscopy in a mouse model of IAV infection, we observed profound neutrophil recruitment, platelet aggregation, neutrophil extracellular trap (NET) production and thrombin activation within the lung microvasculature in vivo. Importantly, deficiency or antagonism of the protease-activated receptor 4 (PAR4) reduced platelet aggregation, NET production, and neutrophil recruitment. Critically, inhibition of thrombin or PAR4 protected mice from virus-induced lung tissue damage and edema. Together, these data imply thrombin-stimulated platelets play a critical role in the activation/recruitment of neutrophils, NET release and directly contribute to IAV pathogenesis in the lung.


Assuntos
Transtornos da Coagulação Sanguínea/imunologia , Plaquetas/imunologia , Armadilhas Extracelulares/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Transtornos da Coagulação Sanguínea/metabolismo , Transtornos da Coagulação Sanguínea/virologia , Plaquetas/metabolismo , Plaquetas/virologia , Modelos Animais de Doenças , Armadilhas Extracelulares/metabolismo , Armadilhas Extracelulares/virologia , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/imunologia , Influenza Humana/metabolismo , Influenza Humana/virologia , Pulmão/metabolismo , Pulmão/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/virologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Agregação Plaquetária/imunologia
6.
Mol Ther Methods Clin Dev ; 20: 95-108, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33376758

RESUMO

Adeno-associated viruses (AAVs) are emerging as one of the vehicles of choice for gene therapy. However, the potential immunogenicity of these vectors is a major limitation of their use, leading to the necessity of a better understanding of how viral vectors engage the innate immune system. In this study, we demonstrate the immune response mediated by an AAV vector in a mouse model. Mice were infected intravenously with 4 × 1012 copies (cp)/kg of AAV8, and the ensuing immune response was analyzed using intravital microscopy during a period of weeks. Administration of AAV8 resulted in the infection of hepatocytes, and this infection led to a moderate, but significant, activation of the immune system in the liver. This host immune response involved platelet aggregation, neutrophil extracellular trap (NET) formation, and the recruitment of monocytes, B cells, and T cells. The resident liver macrophage population, Kupffer cells, was necessary to initiate this immune response, as its depletion abrogated platelet aggregation and NET formation and delayed the recruitment of immune cells. Moreover, the death of liver cells produced by this AAV was moderate and failed to result in a robust, sustained inflammatory response. Altogether, these data suggest that AAV8 is a suitable vector for gene therapy approaches.

7.
Cell ; 183(1): 110-125.e11, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32888431

RESUMO

During respiration, humans breathe in more than 10,000 liters of non-sterile air daily, allowing some pathogens access to alveoli. Interestingly, alveoli outnumber alveolar macrophages (AMs), which favors alveoli devoid of AMs. If AMs, like most tissue macrophages, are sessile, then this numerical advantage would be exploited by pathogens unless neutrophils from the blood stream intervened. However, this would translate to omnipresent persistent inflammation. Developing in vivo real-time intravital imaging of alveoli revealed AMs crawling in and between alveoli using the pores of Kohn. Importantly, these macrophages sensed, chemotaxed, and, with high efficiency, phagocytosed inhaled bacterial pathogens such as P. aeruginosa and S. aureus, cloaking the bacteria from neutrophils. Impairing AM chemotaxis toward bacteria induced superfluous neutrophil recruitment, leading to inappropriate inflammation and injury. In a disease context, influenza A virus infection impaired AM crawling via the type II interferon signaling pathway, and this greatly increased secondary bacterial co-infection.


Assuntos
Bactérias/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Animais , Feminino , Homeostase , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Neutrófilos/imunologia , Fagocitose/imunologia , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/patogenicidade , Alvéolos Pulmonares , Transdução de Sinais , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade
8.
J Exp Med ; 217(4)2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-31978220

RESUMO

Every day, megakaryocytes produce billions of platelets that circulate for several days and eventually are cleared by the liver. The exact removal mechanism, however, remains unclear. Loss of sialic acid residues is thought to feature in the aging and clearance of platelets. Using state-of-the-art spinning disk intravital microscopy to delineate the different compartments and cells of the mouse liver, we observed rapid accumulation of desialylated platelets predominantly on Kupffer cells, with only a few on endothelial cells and none on hepatocytes. Kupffer cell depletion prevented the removal of aged platelets from circulation. Ashwell-Morell receptor (AMR) deficiency alone had little effect on platelet uptake. Macrophage galactose lectin (MGL) together with AMR mediated clearance of desialylated or cold-stored platelets by Kupffer cells. Effective clearance is critical, as mice with an aged platelet population displayed a bleeding phenotype. Our data provide evidence that the MGL of Kupffer cells plays a significant role in the removal of desialylated platelets through a collaboration with the AMR, thereby maintaining a healthy and functional platelet compartment.


Assuntos
Assialoglicoproteínas/metabolismo , Plaquetas/metabolismo , Galactose/metabolismo , Células de Kupffer/metabolismo , Lectinas Tipo C/metabolismo , Proteínas de Membrana/metabolismo , Fagocitose , Animais , Anticorpos/imunologia , Assialoglicoproteínas/imunologia , Células Cultivadas , Voluntários Saudáveis , Humanos , Lectinas Tipo C/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo
9.
Blood ; 135(15): 1281-1286, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-31951648

RESUMO

Antiplatelet therapies have been proposed for the treatment of sepsis, a syndrome resulting from a dysregulated immune response and inappropriate activation of coagulation. Acetylsalicylic acid (ASA) may serve as a potential therapeutic strategy to prevent infection-induced coagulopathy and associated tissue damage. Using intravital microscopy, we found that Staphylococcus aureus infection induced neutrophil recruitment, platelet aggregation, and neutrophil extracellular trap (NET) release in the liver. Mice pretreated with ASA, or animals receiving ASA 3 hours postinfection, had significantly reduced platelet aggregation and NET release. Additionally, ASA-treated mice had reduced intravascular thrombin activity and microvascular occlusion as compared with untreated S aureus-infected mice. This inhibition of coagulation was accompanied by decreased levels of alanine aminotransferase and aspartate aminotransferase in the plasma, indicating less liver damage. Finally, bacterial loads (colony-forming units per milliliter) in liver, lung, and spleen were not different between groups, and the phagocytic capacity of Kupffer cells was preserved following ASA treatment. These results suggest that ASA may serve as a therapeutic approach to sepsis through its ability to reduce the deleterious action of immunothrombi while maintaining innate immune functions.


Assuntos
Aspirina/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Intravascular Disseminada/prevenção & controle , Inibidores da Agregação Plaquetária/uso terapêutico , Sepse/complicações , Infecções Estafilocócicas/complicações , Animais , Coagulação Intravascular Disseminada/sangue , Coagulação Intravascular Disseminada/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Agregação Plaquetária/efeitos dos fármacos , Sepse/sangue , Infecções Estafilocócicas/sangue , Staphylococcus aureus/fisiologia
10.
Platelets ; 31(7): 925-931, 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-31851856

RESUMO

After infection, neutrophils release neutrophil extracellular traps (NETs), decondensed DNA fibers decorated with both nuclear proteins and proteins derived from intracellular granules. These structures have a fundamental role in the development of immunothrombosis; a physiological process mediated by immune cells and molecules from the coagulation system that facilitates the recognition, containment, and destruction of pathogens. Although NETs and immunothrombi are widely hypothesized to be key host defense responses responsible for limiting bacterial dissemination, their actual role in this process has not been formally assessed within the context of a bloodstream infection. Mice were first treated with LPS to generate inflammation (NETs and immunothrombi) and then bacteria dissemination was analyzed by intravital microscopy and colony-forming units (CFU) assay. Blocking NETs or coagulation by the administration of DNase or Argatroban (thrombin inhibitor), respectively, did not modify the percentage of bacteria capture by Kupffer cells, neutrophils or platelets. Moreover, both inhibitors reduced the number of bacteria in the spleen, without modifying CFUs in the liver or lung. In conclusion, we demonstrate that immunothrombi are not necessary to limit the dissemination of bloodstream bacterial infections.


Assuntos
Imunoterapia/métodos , Sepse/tratamento farmacológico , Animais , Modelos Animais de Doenças , Humanos , Camundongos
11.
Cell Rep ; 28(4): 896-908.e5, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31340152

RESUMO

We investigated the contribution of human platelets to macrophage effector properties in the presence of lipopolysaccharide (LPS), as well as the beneficial effects and time frame for platelet transfusion in septic animals. Our results show that platelets sequester both pro-(TNF-α/IL-6) and anti-(IL-10) inflammatory cytokines released by monocytes. Low LPS concentrations (0.01 ng/mL) induced M2 macrophage polarization by decreasing CD64 and augmenting CD206 and CD163 expression; yet, the presence of platelets skewed monocytes toward type 1 macrophage (M1) phenotype in a cell-contact-dependent manner by the glycoprotein Ib (GPIb)-CD11b axis. Accordingly, platelet-licensed macrophages showed increased TNF-α levels, bacterial phagocytic activity, and a reduced healing capability. Platelet transfusion increased inducible nitric oxide synthase (iNOS)+ macrophages, improving bacterial clearance and survival rates in septic mice up to 6 h post-infection, an effect that was abolished by CD11b and GPIb blockade. Our results demonstrate that platelets orchestrate macrophage effector responses, improving the clinical outcome of sepsis in a narrow but relevant time frame.


Assuntos
Plaquetas/metabolismo , Polaridade Celular , Inflamação/patologia , Macrófagos/patologia , Sepse/sangue , Animais , Anti-Inflamatórios/metabolismo , Plaquetas/efeitos dos fármacos , Antígeno CD11b/metabolismo , Comunicação Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fenótipo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Transfusão de Plaquetas , Sepse/patologia , Choque Séptico/patologia , Análise de Sobrevida
12.
Front Immunol ; 10: 2988, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31969883

RESUMO

Non-alcoholic fatty liver disease is a spectrum of liver pathology ranging from simple steatosis to steatohepatitis and can progress to diseases associated with poor outcomes including cirrhosis and hepatocellular carcinoma (HCC). NAFLD research has typically focused on the pathophysiology associated with lipid metabolism, using traditional measures such as histology and serum transaminase assessment; these methods have provided key information regarding NAFLD progression. Although valuable, these techniques are limited in providing further insight into the mechanistic details of inflammation associated with NAFLD. Intravital microscopy (IVM) is an advanced tool that allows for real-time visualization of cellular behavior and interaction in a living animal. Extensive IVM imaging has been conducted in liver, but, in the context of NAFLD, this technique has been regularly avoided due to significant tissue autofluorescence, a phenomenon that is exacerbated with steatosis. Here, we demonstrate that, using multiple imaging platforms and optimization techniques to minimize autofluorescence, IVM in fatty liver is possible. Successful fatty liver intravital imaging provides details on cell trafficking, recruitment, function, and behavior in addition to information about blood flow and vessel dynamics, information which was previously difficult to obtain. As more than 30% of the global population is overweight/obese, there is a significant proportion of the population at risk for NAFLD and complications due to NAFLD (liver decompensation, cirrhosis, HCC). IVM has the potential to elucidate the poorly understood mechanisms surrounding liver inflammation and NAFLD progression and possesses the potential to identify key processes that may be targeted for future therapeutic interventions in NAFLD patients.


Assuntos
Microscopia Intravital , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Rastreamento de Células , Modelos Animais de Doenças , Progressão da Doença , Imunofluorescência , Imuno-Histoquímica , Microscopia Intravital/métodos , Camundongos , Fenótipo
13.
Clin Immunol ; 183: 254-262, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28863968

RESUMO

NETosis is a host defense mechanism associated with inflammation and tissue damage. Experimental models show that platelets and von Willebrand factor (VWF) are key elements for intravascular NETosis. We determined NETosis in septic and burn patients at 1 and 4days post-admission (dpa). Nucleosomes were elevated in patients. In septics, they correlated with Human Neutrophil Elastase (HNE)-DNA complexes and SOFA score at 1dpa, and were associated with mortality. Patient's neutrophils had spontaneous NETosis and were unresponsive to stimulation. Although platelet P-selectin and TNF-α were increased in both groups, higher platelet TLR-4 expression, VWF levels and IL-6 were found in septics at 1dpa. Neither platelet activation markers nor cytokines correlated with nucleosomes or HNE-DNA. Nucleosomes could be indicators of organ damage and predictors of mortality in septic but not in burn patients. Platelet activation, VWF and cytokines do not appear to be key mediators of NETosis in these patient groups.


Assuntos
Queimaduras/metabolismo , Armadilhas Extracelulares/fisiologia , Nucleossomos/metabolismo , Sepse/metabolismo , Adulto , Idoso , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Ativação Plaquetária , Fator de von Willebrand
14.
Sci Rep ; 6: 38738, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27958278

RESUMO

The mechanisms underlying increased thrombotic risk in chronic myeloproliferative neoplasms (MPN) are incompletely understood. We assessed whether neutrophil extracellular traps (NETs), which promote thrombosis, contribute to the procoagulant state in essential thrombocythemia, polycythemia vera and myelofibrosis (MF) patients. Although MPN neutrophils showed increased basal reactive oxygen species (ROS), enhanced NETosis by unstimulated neutrophils was an infrequent finding, whereas PMA-triggered NETosis was impaired, particularly in MF, due to decreased PMA-triggered ROS production. Elevated circulating nucleosomes were a prominent finding and were higher in patients with advanced disease, which may have potential prognostic implication. Histone-MPO complexes, proposed as specific NET biomarker, were seldomly detected, suggesting NETs may not be the main source of nucleosomes in most patients, whereas their correlation with high LDH points to increased cell turn-over as a plausible origin. Lack of association of nucleosomes or NETs with thrombosis or activation markers does not support their use as predictors of thrombosis although prospective studies in a larger cohort may help define their potential contribution to MPN thrombosis. These results do not provide evidence for relevant in vivo NETosis in MPN patients under steady state conditions, although availability of standardized NET biomarkers may contribute to further research in this field.


Assuntos
Biomarcadores Tumorais/sangue , Armadilhas Extracelulares/metabolismo , Neoplasias Hematológicas/sangue , Transtornos Mieloproliferativos/sangue , Nucleossomos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença Crônica , Feminino , Neoplasias Hematológicas/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Mieloproliferativos/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Peroxidase/sangue , Espécies Reativas de Oxigênio/sangue
15.
PLoS One ; 11(12): e0168647, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28005949

RESUMO

INTRODUCTION AND OBJECTIVE: Diabetes is characterized by chronic inflammation, endothelial dysfunction, increased risk of infections and early cardiovascular disease. By releasing neutrophil extracellular traps (NETs), neutrophils kill bacteria and exert pro-inflammatory and pro-thrombotic activities. Increased NETosis has been found in cross-sectional studies including treated type 2 diabetes mellitus (T2DM) patients. In this study, we determined whether the ability of neutrophils to form NETs differs in diabetic patients pre- and post-hyperglycemic control versus healthy donors (HD), and the relationship between NETosis with pro-thrombotic, pro-inflammatory biomarkers and thrombotic clinical events. METHODS: Diabetic patients recently diagnosed and after 6 and 12 months of treatment (N = 25) and HD (N = 25) were included. NET formation was studied by microscopy and fluorometry. Nucleosomes, HNE-DNA complexes, von Willebrand factor (vWF), IL6 and TNFα plasma levels were measured by ELISA and P-selectin on the platelet surface was assessed by cytometry. RESULTS: Basal levels of NETs in recently diagnosed T2DM patients were higher compared to HD. While TNFα stimulation of control neutrophils resulted in DNA release, patient neutrophils were not responsive. Although glycemia decreased after 6 months of metformin treatment, basal and TNFα and PMA-stimulated NETs reached normal values after 12 months. Compared to controls, nucleosomes, HNE-DNA complexes, IL-6 and TNFα levels were increased in recently diagnosed patients and decreased after 12 months of treatment. P-selectin and vWF levels were similar in both populations. CONCLUSION: Our data suggest that NETs could represent a biomarker for T2DM. Increased NETosis in T2DM patients does not appear to be the consequence of impaired glycemic control but rather due to pro-inflammatory cytokines and is not related to thrombotic events.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Armadilhas Extracelulares/metabolismo , Glucose/metabolismo , Hiperglicemia/tratamento farmacológico , Hipoglicemiantes/farmacologia , Inflamação/epidemiologia , Plaquetas/metabolismo , Estudos de Casos e Controles , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Armadilhas Extracelulares/efeitos dos fármacos , Feminino , Humanos , Hiperglicemia/etiologia , Hiperglicemia/metabolismo , Inflamação/prevenção & controle , Masculino , Pessoa de Meia-Idade
16.
Front Immunol ; 7: 271, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27458459

RESUMO

In addition to being key elements in hemostasis and thrombosis, platelets have an important role in the inflammatory and innate immune response. This activity is associated with their capability to recognize pathogens through the expression of toll-like receptors, the secretion of various cytokines, chemokines, and growth factors stored within their granules, and the expression of cell adhesion molecules that allows interaction with other immune cells, mainly neutrophils and monocytes. As part of the first line of defense, neutrophils control invading pathogens by phagocytosis, the release of antimicrobial proteins during degranulation, or through the formation of web-like structures named neutrophil extracellular traps (NETs). NETs are formed by chromatin, proteases, and antimicrobial proteins, and their main function is to trap and kill bacteria, virus, and fungi, avoiding their dissemination. Besides microorganisms, NET formation is also triggered by proinflammatory molecules and platelets. The uncontrolled formation of NETs might exert tissue damage and has been involved in a pathogenic mechanism of autoimmune and prothrombotic clinical conditions. In this review, we discuss the role of platelets in NET generation highlighting the mediators, stimuli, and molecular mechanisms involved in this phenomenon, both in human and murine models.

17.
Acta bioquím. clín. latinoam ; 50(2): 247-254, jun. 2016.
Artigo em Espanhol | LILACS | ID: biblio-837603

RESUMO

Además de ser elementos clave en la hemostasia y trombosis, las plaquetas tienen un rol preponderante en la respuesta inflamatoria e inmune asociada con su capacidad para reconocer patógenos a través de la expresión de los receptores tipo Toll, la secreción de una amplia variedad de citoquinas, quemoquinas y factores de crecimiento almacenados en sus gránulos y por la expresión de moléculas de adhesión que permiten la interacción con otras células vasculares. Como parte de la primera línea de defensa, los neutrófilos controlan la infección por fagocitosis, liberación de proteínas antimicrobianas durante la degranulación o a través de la formación de estructuras tipo red, conocidas como trampas extracelulares de neutrófilos (NETs). Estas están formadas por cromatina, proteasas y proteínas antimicrobianas cuya función principal es atrapar y eliminar bacterias, virus y hongos, impidiendo su diseminación. Además de microorganismos, la formación de NETs es gatillada por moléculas proinflamatorias y plaquetas. Su formación descontrolada puede ocasionar daño tisular y es considerada un mecanismo patogénico de eventos protrombóticos, inflamatorios y autoinmunes. En esta revisión se discute el rol de las plaquetas en la formación de NETs y se destacan los mediadores, estímulos y mecanismos moleculares participantes de este fenómeno, en humanos y modelos murinos.


In addition to being key elements in hemostasis and thrombosis, platelets have an important role in inflammatory and innate immune responses. This activity is associated with their capability to recognize pathogens through the expression of TLRs, the secretion of a wide variety of cytokines, chemokines and growth factors stored within their granules and cell adhesion molecule expresssion that enable interaction with other vascular cells. As part of the first line of defense, neutrophils control invading pathogens by phagocytosis, the release of antimicrobial proteins during degranulation or through the formation of web-like structures known as neutrophil extracellular traps (NETs). NETs are formed by chromatin, proteases and antimicrobial proteins and their main function is to trap and kill bacteria, virus and fungi, thus avoiding their dissemination. Besides microorganisms, NETs formation is also triggered by proinflammatory molecules, and platelets. The uncontrolled formation of NETs might exert tissue damage and has been involved as a pathogenic mechanism of autoimmune and prothrombotic events. In this review, the role of platelets in NET generation is discussed, highlighting the mediators, stimuli and molecular mechanisms involved in this phenomenon, both in human and murine models.


Além de serem elementos chave na hemostasia e trombose, as plaquetas têm um papel preponderante na resposta inflamatória e imune associada a sua capacidade para reconhecer patógenos através da expressão dos receptores tipo Toll, a secreção de uma ampla variedade de citocinas, quemocinas e fatores de crescimento armazenados em seus grânulos e pela expressão de moléculas de adesão que permitem a interação com outras células vasculares. Como parte da primeira linha de defesa, os neutrófilos controlam a infecção por fagocitose, liberação de proteínas antimicrobianas durante a degranulação ou através da formação de estruturas tipo rede, conhecidas como armadilhas extracelulares de neutrófilos (NETs). Elas estão formadas por cromatina, proteases e proteínas antimicrobianas cuja função principal é prender e eliminar bactérias, vírus e fungos, impedindo sua disseminação. Além de microorganismos, a formação de NETs é disparada por moléculas pró-inflamatórias e plaquetas. Sua formação descontrolada pode provocar dano tissular e é considerada um mecanismo patogênico de eventos pró-trombóticos, inflamatórios e autoimunes. Nesta revisão é discutido o papel das plaquetas na formação de NETs, destacando os mediadores, estímulos e mecanismos moleculares participantes deste fenômeno, em humanos e modelos murídeos.


Assuntos
Humanos , Plaquetas , Armadilhas Extracelulares , Neutrófilos , Trombose , Homeostase
18.
J Leukoc Biol ; 99(1): 153-62, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26320263

RESUMO

In addition to being key elements in hemostasis and thrombosis, platelets amplify neutrophil function. We aimed to gain further insight into the stimuli, mediators, molecular pathways, and regulation of neutrophil extracellular trap formation mediated by human platelets. Platelets stimulated by lipopolysaccharide, a wall component of gram-negative bacteria, Pam3-cysteine-serine-lysine 4, a mimetic of lipopeptide from gram-positive bacteria, Escherichia coli, Staphylococcus aureus, or physiologic platelet agonists promoting neutrophil extracellular trap formation and myeloperoxidase-associated DNA activity under static and flow conditions. Although P-selectin or glycoprotein IIb/IIIa were not involved, platelet glycoprotein Ib, neutrophil cluster of differentiation 18, and the release of von Willebrand factor and platelet factor 4 seemed to be critical for the formation of neutrophil extracellular traps. The secretion of these molecules depended on thromboxane A(2) production triggered by lipopolysaccharide or Pam3-cysteine-serine-lysine 4 but not on high concentrations of thrombin. Accordingly, aspirin selectively inhibited platelet-mediated neutrophil extracellular trap generation. Signaling through extracellular signal-regulated kinase, phosphatidylinositol 3-kinase, and Src kinases, but not p38 or reduced nicotinamide adenine dinucleotide phosphate oxidase, was involved in platelet-triggered neutrophil extracellular trap release. Platelet-mediated neutrophil extracellular trap formation was inhibited by prostacyclin. Our results support a role for stimulated platelets in promoting neutrophil extracellular trap formation, reveal that an endothelium-derived molecule contributes to limiting neutrophil extracellular trap formation, and highlight platelet inhibition as a potential target for controlling neutrophil extracellular trap cell death.


Assuntos
Plaquetas/metabolismo , Armadilhas Extracelulares/imunologia , Armadilhas Extracelulares/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Ativação Plaquetária , Transdução de Sinais , Células Endoteliais/metabolismo , Humanos , Lipopeptídeos/imunologia , Lipopolissacarídeos/imunologia , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/imunologia , Receptores de Superfície Celular/metabolismo
19.
PLoS Negl Trop Dis ; 9(7): e0003927, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26161745

RESUMO

NETosis is a process by which neutrophils extrude their DNA together with bactericidal proteins that trap and/or kill pathogens. In the present study, we evaluated the ability of Leptospira spp. to induce NETosis using human ex vivo and murine in vivo models. Microscopy and fluorometric studies showed that incubation of human neutrophils with Leptospira interrogans serovar Copenhageni strain Fiocruz L1-130 (LIC) resulted in the release of DNA extracellular traps (NETs). The bacteria number, pathogenicity and viability were relevant factors for induction of NETs, but bacteria motility was not. Entrapment of LIC in the NETs resulted in LIC death; however, pathogenic but not saprophytic Leptospira sp. exerted nuclease activity and degraded DNA. Mice infected with LIC showed circulating NETs after 2 days post-infection (dpi). Depletion of neutrophils with mAb1A8 significantly reduced the amount of intravascular NETs in LIC-infected mice, increasing bacteremia at 3 dpi. Although there was a low bacterial burden, scarce neutrophils and an absence of inflammation in the early stages of infection in the kidney and liver, at the beginning of the leptospiruric phase, the bacterial burden was significantly higher in kidneys of neutrophil-depleted-mice compared to non-depleted and infected mice. Surprisingly, interstitial nephritis was of similar intensity in both groups of infected mice. Taken together, these data suggest that LIC triggers NETs, and that the intravascular formation of these DNA traps appears to be critical not only to prevent early leptospiral dissemination but also to preclude further bacterial burden.


Assuntos
Armadilhas Extracelulares/imunologia , Leptospira/fisiologia , Leptospirose/imunologia , Neutrófilos/imunologia , Animais , Humanos , Imunidade Inata , Leptospira/imunologia , Leptospirose/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/microbiologia
20.
Glycobiology ; 24(10): 966-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24957054

RESUMO

Galectins (Gals), a family of mammalian lectins, play diverse roles under physiological and pathological conditions. Here, we analyzed the tandem-repeat Gal-8 synthesis, secretion and effects on the endothelium physiology. Gal-8M and Gal-8L isoforms were secreted under basal conditions by human microvascular endothelial cells (HMEC-1). However, expression and secretion of the Gal-8M isoform, but not Gal-8L, were increased in response to bacterial lipopolysaccharide (LPS) stimulus and returned to control values after LPS removal. Similarly, cell surface Gal-8 exposure was increased after stimulation with LPS. To evaluate Gal-8 effects on the endothelium physiology, HMEC-1 cells were incubated in the presence of recombinant Gal-8M. Pretreated HMEC-1 cells became proadhesive to human normal platelets, indicating that Gal-8 actually activates endothelial cells. This effect was specific for lectin activity as it was prevented by the simultaneous addition of lactose, but not by sucrose. Endothelial cells also increased their exposition of von Willebrand factor after Gal-8 treatment, which constitutes another feature of cell activation that could be, in turn, responsible for the observed platelet adhesion. Several pro-inflammatory molecules were abundantly produced by Gal-8 stimulated endothelial cells: CXCL1 (GRO-α), GM-CSF, IL-6 and CCL5 (RANTES), and in a lower degree CCL2 (MCP-1), CXCL3 (GRO-γ) and CXCL8 (IL-8). In agreement, Gal-8M induced nuclear factor kappa B phosphorylation. Altogether, these results not only confirm the pro-inflammatory role we have already proposed for Gal-8 in other cellular systems but also suggest that this lectin is orchestrating the interaction between leukocytes, platelets and endothelial cells.


Assuntos
Endotélio/metabolismo , Galectinas/metabolismo , Interleucina-8/metabolismo , Galectinas/genética , Humanos , Interleucina-8/genética , Lipopolissacarídeos/química , Sequências de Repetição em Tandem/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA