Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.618
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(23): 5740-5758.e17, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34735796

RESUMEN

Biofilms are community architectures adopted by bacteria inclusive of a self-formed extracellular matrix that protects resident bacteria from diverse environmental stresses and, in many species, incorporates extracellular DNA (eDNA) and DNABII proteins for structural integrity throughout biofilm development. Here, we present evidence that this eDNA-based architecture relies on the rare Z-form. Z-form DNA accumulates as biofilms mature and, through stabilization by the DNABII proteins, confers structural integrity to the biofilm matrix. Indeed, substances known to drive B-DNA into Z-DNA promoted biofilm formation whereas those that drive Z-DNA into B-DNA disrupted extant biofilms. Importantly, we demonstrated that the universal bacterial DNABII family of proteins stabilizes both bacterial- and host-eDNA in the Z-form in situ. A model is proposed that incorporates the role of Z-DNA in biofilm pathogenesis, innate immune response, and immune evasion.


Asunto(s)
Bacterias/genética , Biopelículas , ADN Bacteriano/química , Matriz Extracelular/metabolismo , Espacio Extracelular/química , Animales , Especificidad de Anticuerpos , Proteínas Bacterianas/metabolismo , Línea Celular , Chinchilla , ADN Cruciforme , Desoxirribonucleasas/metabolismo , Trampas Extracelulares/metabolismo , Humanos , Acetato de Tetradecanoilforbol/farmacología
2.
Nat Immunol ; 24(4): 575-584, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36959290

RESUMEN

Neutrophils are formidable defenders. Their vast numbers, constant production, high cytotoxicity and capacity to produce extracellular traps, underlie their ability to efficiently protect in a microorganism-rich world. However, neutrophils are much more than immune sentinels, as evidenced by the expanding repertoire of functions discovered in the context of tissue homeostasis, regeneration or chronic pathologies. In this Perspective, we discuss general functional features of the neutrophil compartment that may be relevant in most, if not all, physiological scenarios in which they participate, including specialization in naïve tissues, transcriptional noise in the bloodstream as a potential strategy for diversification and functional bias in inflammatory sites. We intentionally present the reader with more questions than answers and propose models and approaches that we hope will shed new light onto the biology of these fascinating cells and spark new directions of research.


Asunto(s)
Trampas Extracelulares , Neutrófilos , Homeostasis
3.
Annu Rev Cell Dev Biol ; 36: 191-218, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32663035

RESUMEN

Neutrophils are critical to innate immunity, including host defense against bacterial and fungal infections. They achieve their host defense role by phagocytosing pathogens, secreting their granules full of cytotoxic enzymes, or expelling neutrophil extracellular traps (NETs) during the process of NETosis. NETs are weblike DNA structures decorated with histones and antimicrobial proteins released by activated neutrophils. Initially described as a means for neutrophils to neutralize pathogens, NET release also occurs in sterile inflammation, promotes thrombosis, and can mediate tissue damage. To effectively manipulate this double-edged sword to fight a particular disease, researchers must work toward understanding the mechanisms driving NETosis. Such understanding would allow the generation of new drugs to promote or prevent NETosis as needed. While knowledge regarding the (patho)physiological roles of NETosis is accumulating, little is known about the cellular and biophysical bases of this process. In this review, we describe and discuss our current knowledge of the molecular, cellular, and biophysical mechanisms mediating NET release as well as open questions in the field.


Asunto(s)
Trampas Extracelulares/metabolismo , Animales , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Citosol/metabolismo , ADN/metabolismo , Humanos
4.
Nat Immunol ; 21(2): 135-144, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932813

RESUMEN

The antimicrobial functions of neutrophils are facilitated by a defensive armamentarium of proteins stored in granules, and by the formation of neutrophil extracellular traps (NETs). However, the toxic nature of these structures poses a threat to highly vascularized tissues, such as the lungs. Here, we identified a cell-intrinsic program that modified the neutrophil proteome in the circulation and caused the progressive loss of granule content and reduction of the NET-forming capacity. This program was driven by the receptor CXCR2 and by regulators of circadian cycles. As a consequence, lungs were protected from inflammatory injury at times of day or in mouse mutants in which granule content was low. Changes in the proteome, granule content and NET formation also occurred in human neutrophils, and correlated with the incidence and severity of respiratory distress in pneumonia patients. Our findings unveil a 'disarming' strategy of neutrophils that depletes protein stores to reduce the magnitude of inflammation.


Asunto(s)
Ritmo Circadiano/inmunología , Inflamación/metabolismo , Neutrófilos/metabolismo , Neumonía/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Animales , Degranulación de la Célula/inmunología , Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/metabolismo , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Humanos , Inflamación/inmunología , Ratones , Neutrófilos/inmunología , Neumonía/complicaciones , Neumonía/inmunología , Proteoma/inmunología , Proteoma/metabolismo , Síndrome de Dificultad Respiratoria/inmunología
5.
Immunity ; 56(12): 2755-2772.e8, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38039967

RESUMEN

In triple-negative breast cancer (TNBC), stromal restriction of CD8+ T cells associates with poor clinical outcomes and lack of responsiveness to immune-checkpoint blockade (ICB). To identify mediators of T cell stromal restriction, we profiled murine breast tumors lacking the transcription factor Stat3, which is commonly hyperactive in breast cancers and promotes an immunosuppressive tumor microenvironment. Expression of the cytokine Chi3l1 was decreased in Stat3-/- tumors. CHI3L1 expression was elevated in human TNBCs and other solid tumors exhibiting T cell stromal restriction. Chi3l1 ablation in the polyoma virus middle T (PyMT) breast cancer model generated an anti-tumor immune response and delayed mammary tumor onset. These effects were associated with increased T cell tumor infiltration and improved response to ICB. Mechanistically, Chi3l1 promoted neutrophil recruitment and neutrophil extracellular trap formation, which blocked T cell infiltration. Our findings provide insight into the mechanism underlying stromal restriction of CD8+ T cells and suggest that targeting Chi3l1 may promote anti-tumor immunity in various tumor types.


Asunto(s)
Trampas Extracelulares , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Linfocitos T CD8-positivos , Línea Celular Tumoral , Citocinas , Trampas Extracelulares/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral
6.
Immunity ; 56(5): 1013-1026.e6, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36944334

RESUMEN

Sepsis is a dysregulated inflammatory consequence of systemic infection. As a result, excessive platelet activation leads to thrombosis and coagulopathy, but we currently lack sufficient understanding of these processes. Here, using the cecal ligation and puncture (CLP) model of sepsis, we observed septic thrombosis and neutrophil extracellular trap formation (NETosis) within the mouse vasculature by intravital microscopy. STING activation in platelets was a critical driver of sepsis-induced pathology. Platelet-specific STING deficiency suppressed platelet activation and granule secretion, which alleviated sepsis-induced intravascular thrombosis and NETosis in mice. Mechanistically, sepsis-derived cGAMP promoted the binding of STING to STXBP2, the assembly of SNARE complex, granule secretion, and subsequent septic thrombosis, which probably depended on the palmitoylation of STING. We generated a peptide, C-ST5, to block STING binding to STXBP2. Septic mice treated with C-ST5 showed reduced thrombosis. Overall, platelet activation via STING reveals a potential strategy for limiting life-threatening sepsis-mediated coagulopathy.


Asunto(s)
Trampas Extracelulares , Sepsis , Trombosis , Animales , Ratones , Plaquetas/metabolismo , Trampas Extracelulares/metabolismo , Ratones Endogámicos C57BL , Proteínas Munc18/metabolismo , Activación Plaquetaria , Sepsis/metabolismo , Trombosis/metabolismo
7.
Nat Immunol ; 20(11): 1444-1455, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591573

RESUMEN

Low exposure to microbial products, respiratory viral infections and air pollution are major risk factors for allergic asthma, yet the mechanistic links between such conditions and host susceptibility to type 2 allergic disorders remain unclear. Through the use of single-cell RNA sequencing, we characterized lung neutrophils in mice exposed to a pro-allergic low dose of lipopolysaccharide (LPS) or a protective high dose of LPS before exposure to house dust mites. Unlike exposure to a high dose of LPS, exposure to a low dose of LPS instructed recruited neutrophils to upregulate their expression of the chemokine receptor CXCR4 and to release neutrophil extracellular traps. Low-dose LPS-induced neutrophils and neutrophil extracellular traps potentiated the uptake of house dust mites by CD11b+Ly-6C+ dendritic cells and type 2 allergic airway inflammation in response to house dust mites. Neutrophil extracellular traps derived from CXCR4hi neutrophils were also needed to mediate allergic asthma triggered by infection with influenza virus or exposure to ozone. Our study indicates that apparently unrelated environmental risk factors can shape recruited lung neutrophils to promote the initiation of allergic asthma.


Asunto(s)
Contaminantes Atmosféricos/inmunología , Alérgenos/inmunología , Asma/inmunología , Trampas Extracelulares/metabolismo , Neutrófilos/inmunología , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Exposición a Riesgos Ambientales/efectos adversos , Trampas Extracelulares/inmunología , Femenino , Humanos , Lipopolisacáridos/inmunología , Pulmón/citología , Pulmón/inmunología , Ratones , Neutrófilos/metabolismo , Orthomyxoviridae/inmunología , Ozono/inmunología , Pyroglyphidae/inmunología , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Regulación hacia Arriba
8.
Immunity ; 55(2): 224-236.e5, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34995475

RESUMEN

During gram-negative septicemia, interactions between platelets and neutrophils initiate a detrimental feedback loop that sustains neutrophil extracellular trap (NET) induction, disseminated intravascular coagulation, and inflammation. Understanding intracellular pathways that control platelet-neutrophil interactions is essential for identifying new therapeutic targets. Here, we found that thrombin signaling induced activation of the transcription factor NFAT in platelets. Using genetic and pharmacologic approaches, as well as iNFATuation, a newly developed mouse model in which NFAT activation can be abrogated in a cell-specific manner, we demonstrated that NFAT inhibition in activated murine and human platelets enhanced their activation and aggregation, as well as their interactions with neutrophils and NET induction. During gram-negative septicemia, NFAT inhibition in platelets promoted disease severity by increasing disseminated coagulation and NETosis. NFAT inhibition also partially restored coagulation ex vivo in patients with hypoactive platelets. Our results define non-transcriptional roles for NFAT that could be harnessed to address pressing clinical needs.


Asunto(s)
Plaquetas/efectos de los fármacos , Factores de Transcripción NFATC/antagonistas & inhibidores , Agregación Plaquetaria/efectos de los fármacos , Sepsis/patología , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Comunicación Celular/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Humanos , Inflamación , Ratones , Factores de Transcripción NFATC/metabolismo , Neutrófilos/metabolismo , Receptores de Trombina/metabolismo , Sepsis/metabolismo
9.
Physiol Rev ; 103(1): 277-312, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35951483

RESUMEN

Neutrophil extracellular trap (NET) formation, first described in 2004 as a previously unknown strategy of neutrophils to fight microbes, has attracted an increasing interest in the research community. NETs are formed when neutrophils externalize their decondensed chromatin together with content from their azurophilic granules. In addition to their role in defense against microbes, NETs have been implicated as mediators of pathology in sterile inflammation, such as cancer and autoimmunity, and their potential as therapeutic targets is actively explored. However, targeting of NETs is challenging since the beneficial effects of their removal need to be balanced against the potential harmful loss of their function in microbial defense. Moreover, depending on the stimuli or species, NETs can be formed via distinct mechanisms and are not always made up of the same components, making direct comparisons between various studies challenging. This review focuses on the role of NETs in cancer-associated pathology, such as thrombosis, organ dysfunction, and metastasis. Different strategies to target NETs, by either preventing their formation or degrading existing ones, are also discussed.


Asunto(s)
Trampas Extracelulares , Neoplasias , Trombosis , Cromatina , Humanos , Neoplasias/patología , Neutrófilos
10.
Immunity ; 52(5): 729-731, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32433945

RESUMEN

Neutrophil extracellular traps (NETs) can promote tumor growth and metastases, but whether NETs impact the tumor immune microenvironment remains underexplored. In this issue of Immunity, Teijeira et al. discover that NETs shield tumor cells from cytotoxic immune cells, resulting in impaired tumor clearance.


Asunto(s)
Trampas Extracelulares , Neoplasias , Quimiocinas , Humanos , Neutrófilos , Microambiente Tumoral
11.
Immunity ; 52(5): 856-871.e8, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32289253

RESUMEN

Neutrophils are expanded and abundant in cancer-bearing hosts. Under the influence of CXCR1 and CXCR2 chemokine receptor agonists and other chemotactic factors produced by tumors, neutrophils, and granulocytic myeloid-derived suppressor cells (MDSCs) from cancer patients extrude their neutrophil extracellular traps (NETs). In our hands, CXCR1 and CXCR2 agonists proved to be the major mediators of cancer-promoted NETosis. NETs wrap and coat tumor cells and shield them from cytotoxicity, as mediated by CD8+ T cells and natural killer (NK) cells, by obstructing contact between immune cells and the surrounding target cells. Tumor cells protected from cytotoxicity by NETs underlie successful cancer metastases in mice and the immunotherapeutic synergy of protein arginine deiminase 4 (PAD4) inhibitors, which curtail NETosis with immune checkpoint inhibitors. Intravital microscopy provides evidence of neutrophil NETs interfering cytolytic cytotoxic T lymphocytes (CTLs) and NK cell contacts with tumor cells.


Asunto(s)
Trampas Extracelulares/metabolismo , Neoplasias Experimentales/terapia , Receptores de Quimiocina/agonistas , Receptores de Interleucina-8A/agonistas , Receptores de Interleucina-8B/agonistas , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Células HT29 , Humanos , Microscopía Intravital/métodos , Células Asesinas Naturales/inmunología , Ligandos , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Receptores de Interleucina-8A/inmunología , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Linfocitos T Citotóxicos/inmunología
12.
Immunity ; 52(4): 700-715.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294409

RESUMEN

The omentum is a visceral adipose tissue rich in fat-associated lymphoid clusters (FALCs) that collects peritoneal contaminants and provides a first layer of immunological defense within the abdomen. Here, we investigated the mechanisms that mediate the capture of peritoneal contaminants during peritonitis. Single-cell RNA sequencing and spatial analysis of omental stromal cells revealed that the surface of FALCs were covered by CXCL1+ mesothelial cells, which we termed FALC cover cells. Blockade of CXCL1 inhibited the recruitment and aggregation of neutrophils at FALCs during zymosan-induced peritonitis. Inhibition of protein arginine deiminase 4, an enzyme important for the release of neutrophil extracellular traps, abolished neutrophil aggregation and the capture of peritoneal contaminants by omental FALCs. Analysis of omental samples from patients with acute appendicitis confirmed neutrophil recruitment and bacterial capture at FALCs. Thus, specialized omental mesothelial cells coordinate the recruitment and aggregation of neutrophils to capture peritoneal contaminants.


Asunto(s)
Apendicitis/inmunología , Linfocitos/inmunología , Neutrófilos/inmunología , Epiplón/inmunología , Peritonitis/inmunología , Células del Estroma/inmunología , Enfermedad Aguda , Animales , Apendicitis/genética , Apendicitis/microbiología , Comunicación Celular/inmunología , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Epitelio/inmunología , Epitelio/microbiología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Trampas Extracelulares/inmunología , Femenino , Expresión Génica , Humanos , Linfocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neutrófilos/microbiología , Epiplón/microbiología , Peritonitis/inducido químicamente , Peritonitis/genética , Peritonitis/microbiología , Arginina Deiminasa Proteína-Tipo 4/genética , Arginina Deiminasa Proteína-Tipo 4/inmunología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Células del Estroma/microbiología , Técnicas de Cultivo de Tejidos , Zimosan/administración & dosificación
13.
Immunity ; 51(3): 413-414, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31533050

RESUMEN

Gallstone formation in adults is a common, yet incompletely understood disease process. In this issue, Muñoz et al. (2019) report a pathogenic link between neutrophil extracellular traps (NETs) and the formation of gallstones.


Asunto(s)
Trampas Extracelulares , Cálculos Biliares , Adulto , Humanos , Neutrófilos
14.
Immunity ; 51(3): 443-450.e4, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31422870

RESUMEN

The presence of gallstones (cholelithiasis) is a highly prevalent and severe disease and one of the leading causes of hospital admissions worldwide. Due to its substantial health impact, we investigated the biological mechanisms that lead to the formation and growth of gallstones. We show that gallstone assembly essentially requires neutrophil extracellular traps (NETs). We found consistent evidence for the presence of NETs in human and murine gallstones and describe an immune-mediated process requiring activation of the innate immune system for the formation and growth of gallstones. Targeting NET formation via inhibition of peptidyl arginine deiminase type 4 or abrogation of reactive oxygen species (ROS) production, as well as damping of neutrophils by metoprolol, effectively inhibit gallstone formation in vivo. Our results show that after the physicochemical process of crystal formation, NETs foster their assembly into larger aggregates and finally gallstones. These insights provide a feasible therapeutic concept to prevent cholelithiasis in patients at risk.


Asunto(s)
Trampas Extracelulares/inmunología , Cálculos Biliares/inmunología , Neutrófilos/inmunología , Animales , Femenino , Humanos , Inmunidad Innata/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/inmunología
15.
Immunol Rev ; 321(1): 263-279, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37712361

RESUMEN

The process of neutrophil extracellular traps (NETs) formation, called NETosis, is a peculiar death modality of neutrophils, which was first observed as an immune response against bacterial infection. However, recent work has revealed the unique biology of NETosis in facilitating tumor metastatic process. Neutrophil extracellular traps released by the tumor microenvironment (TME) shield tumor cells from cytotoxic immunity, leading to impaired tumor clearance. Besides, tumor cells tapped by NETs enable to travel through vessels and subsequently seed distant organs. Targeted ablation of NETosis has been proven to be beneficial in potentiating the efficacy of cancer immunotherapy in the metastatic settings. This review outlines the impact of NETosis at almost all stages of tumor metastasis. Furthermore, understanding the multifaceted interplay between NETosis and the TME components is crucial for supporting the rational development of highly effective combination immunotherapeutic strategies with anti-NETosis for patients with metastatic disease.


Asunto(s)
Trampas Extracelulares , Neoplasias , Humanos , Neutrófilos , Neoplasias/terapia , Neoplasias/patología , Inmunoterapia , Microambiente Tumoral
16.
Immunity ; 49(5): 798-800, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30462996

RESUMEN

In a recent study published in Science, Albrengues et al. (2018) unveil an intriguing mechanism whereby the release of neutrophil extra-cellular traps during chronic lung inflammation awakens dormant malignant cells and contributes to cancer progression.


Asunto(s)
Trampas Extracelulares , Neoplasias , Neumonía , Animales , Inflamación , Ratones , Neutrófilos
17.
Immunol Rev ; 314(1): 13-35, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36527200

RESUMEN

Neutrophils sense microbes and host inflammatory mediators, and traffic to sites of infection where they direct a broad armamentarium of antimicrobial products against pathogens. Neutrophils are also activated by damage-associated molecular patterns (DAMPs), which are products of cellular injury that stimulate the innate immune system through pathways that are similar to those activated by microbes. Neutrophils and platelets become activated by injury, and cluster and cross-signal to each other with the cumulative effect of driving antimicrobial defense and hemostasis. In addition, neutrophil extracellular traps are extracellular chromatin and granular constituents that are generated in response to microbial and damage motifs and are pro-thrombotic and injurious. Although neutrophils can worsen tissue injury, neutrophils may also have a role in facilitating wound repair following injury. A central theme of this review relates to how critical functions of neutrophils that evolved to respond to infection and damage modulate the tumor microenvironment (TME) in ways that can promote or limit tumor progression. Neutrophils are reprogrammed by the TME, and, in turn, can cross-signal to tumor cells and reshape the immune landscape of tumors. Importantly, promising new therapeutic strategies have been developed to target neutrophil recruitment and function to make cancer immunotherapy more effective.


Asunto(s)
Trampas Extracelulares , Neutrófilos , Humanos , Plaquetas/metabolismo , Plaquetas/patología , Células Endoteliales , Inflamación , Linfocitos T , Trampas Extracelulares/metabolismo
18.
Immunol Rev ; 314(1): 376-398, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36560865

RESUMEN

Thrombosis remains a major problem in our society, manifesting across multiple demographic groups and with high associated morbidity and mortality. Thrombus development is the result of a complex mechanism in which multiple cell types and soluble factors play a crucial role. One cell that has gained the most attention in recent years is the neutrophil. This key member of the innate immune system can form neutrophil extracellular traps (NETs) in response to activating stimuli in circulation. NETs form a scaffold for thrombus formation, both initiating the process and stabilizing the final product. As the first responders of the host immune system, neutrophils have the flexibility to recognize a variety of molecules and can quickly interact with a range of different cell types. This trait makes them sensitive to exogenous stimuli. NET formation in response to pathogens is well established, leading to immune-mediated thrombus formation or immunothrombosis. NETs can also be formed during sterile inflammation through the activation of neutrophils by fellow immune cells including platelets, or activated endothelium. In chronic inflammatory settings, NETs can ultimately promote the development of tissue fibrosis, with organ failure as an end-stage outcome. In this review, we discuss the different pathways through which neutrophils can be activated toward NET formation and how these processes can result in a shared outcome: thrombus formation. Finally, we evaluate these different interactions and mechanisms for their potential as therapeutic targets, with neutrophil-targeted therapies providing a future approach to treating thrombosis. In contrast to current practices, such treatment could result in reduced pathogenic blood clot formation without increasing the risk of bleeding.


Asunto(s)
Trampas Extracelulares , Trombosis , Humanos , Trampas Extracelulares/metabolismo , Trombosis/metabolismo , Trombosis/patología , Neutrófilos , Inflamación/metabolismo , Fenotipo
19.
Immunol Rev ; 314(1): 399-412, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36440642

RESUMEN

Coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in more than 6 million deaths worldwide. COVID-19 is a respiratory disease characterized by pulmonary dysfunction leading to acute respiratory distress syndrome (ARDs), as well as disseminated coagulation, and multi-organ dysfunction. Neutrophils and neutrophil extracellular traps (NETs) have been implicated in the pathogenesis of COVID-19. In this review, we highlight key gaps in knowledge, discuss the heterogeneity of neutrophils during the evolution of the disease, how they can contribute to COVID-19 pathogenesis, and potential therapeutic strategies that target neutrophil-mediated inflammatory responses.


Asunto(s)
COVID-19 , Trampas Extracelulares , Humanos , COVID-19/patología , Neutrófilos , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA