Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 142(17): 1463-1477, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37441848

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with high morbidity and mortality. Excessive neutrophil infiltration into the pulmonary airspace is the main cause for the acute inflammation and lung injury. Platelets have been implicated in the pathogenesis of ALI/ARDS, but the underlying mechanisms are not fully understood. Here, we show that the immunoreceptor tyrosine-based activation motif-coupled immunoglobulin-like platelet receptor, glycoprotein VI (GPVI), plays a key role in the early phase of pulmonary thrombo-inflammation in a model of lipopolysaccharide (LPS)-induced ALI in mice. In wild-type (WT) control mice, intranasal LPS application triggered severe pulmonary and blood neutrophilia, hypothermia, and increased blood lactate levels. In contrast, GPVI-deficient mice as well as anti-GPVI-treated WT mice were markedly protected from pulmonary and systemic compromises and showed no increased pulmonary bleeding. High-resolution multicolor microscopy of lung sections and intravital confocal microcopy of the ventilated lung revealed that anti-GPVI treatment resulted in less stable platelet interactions with neutrophils and overall reduced platelet-neutrophil complex (PNC) formation. Anti-GPVI treatment also reduced neutrophil crawling and adhesion on endothelial cells, resulting in reduced neutrophil transmigration and alveolar infiltrates. Remarkably, neutrophil activation was also diminished in anti-GPVI-treated animals, associated with strongly reduced formation of PNC clusters and neutrophil extracellular traps (NETs) compared with that in control mice. These results establish GPVI as a key mediator of neutrophil recruitment, PNC formation, and NET formation (ie, NETosis) in experimental ALI. Thus, GPVI inhibition might be a promising strategy to reduce the acute pulmonary inflammation that causes ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Neumonía , Síndrome de Dificultad Respiratoria , Animales , Ratones , Lesión Pulmonar Aguda/patología , Células Endoteliales/patología , Inflamación/patología , Lipopolisacáridos/efectos adversos , Pulmón/patología , Infiltración Neutrófila , Neutrófilos/patología , Neumonía/patología , Síndrome de Dificultad Respiratoria/patología
2.
Basic Res Cardiol ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38811421

RESUMEN

Neutrophils are not only involved in immune defense against infection but also contribute to the exacerbation of tissue damage after ischemia and reperfusion. We have previously shown that genetic ablation of regulatory Gαi proteins in mice has both protective and deleterious effects on myocardial ischemia reperfusion injury (mIRI), depending on which isoform is deleted. To deepen and analyze these findings in more detail the contribution of Gαi2 proteins in resident cardiac vs circulating blood cells for mIRI was first studied in bone marrow chimeras. In fact, the absence of Gαi2 in all blood cells reduced the extent of mIRI (22,9% infarct size of area at risk (AAR) Gnai2-/- → wt vs 44.0% wt → wt; p < 0.001) whereas the absence of Gαi2 in non-hematopoietic cells increased the infarct damage (66.5% wt → Gnai2-/- vs 44.0% wt → wt; p < 0.001). Previously we have reported the impact of platelet Gαi2 for mIRI. Here, we show that infarct size was substantially reduced when Gαi2 signaling was either genetically ablated in neutrophils/macrophages using LysM-driven Cre recombinase (AAR: 17.9% Gnai2fl/fl LysM-Cre+/tg vs 42.0% Gnai2fl/fl; p < 0.01) or selectively blocked with specific antibodies directed against Gαi2 (AAR: 19.0% (anti-Gαi2) vs 49.0% (IgG); p < 0.001). In addition, the number of platelet-neutrophil complexes (PNCs) in the infarcted area were reduced in both, genetically modified (PNCs: 18 (Gnai2fl/fl; LysM-Cre+/tg) vs 31 (Gnai2fl/fl); p < 0.001) and in anti-Gαi2 antibody-treated (PNCs: 9 (anti-Gαi2) vs 33 (IgG); p < 0.001) mice. Of note, significant infarct-limiting effects were achieved with a single anti-Gαi2 antibody challenge immediately prior to vessel reperfusion without affecting bleeding time, heart rate or cellular distribution of neutrophils. Finally, anti-Gαi2 antibody treatment also inhibited transendothelial migration of human neutrophils (25,885 (IgG) vs 13,225 (anti-Gαi2) neutrophils; p < 0.001), collectively suggesting that a therapeutic concept of functional Gαi2 inhibition during thrombolysis and reperfusion in patients with myocardial infarction should be further considered.

3.
Clin Gastroenterol Hepatol ; 21(2): 541-542.e1, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35123095

RESUMEN

Approximately 20% of patients with ulcerative colitis (UC) who undergo total proctocolectomy (TPC) with ileal pouch-anal anastomosis (IPAA) develop chronic pouch inflammation (CPI).1 Given the involvement of the small bowel in CPI, there is no consensus on whether it should be managed more like UC or Crohn's disease (CD). Despite limited evidence, biologics are often used for CPI with clinical response rates of 20%-60% with adalimumab and infliximab, 50%-80% with ustekinumab, and 30%-70% with vedolizumab.2,3 Earlier biologic therapy has been associated with greater rates of response and favorable long-term outcomes in patients with CD, however not UC.4-7 The impact of earlier initiation of biologic therapy on CPI clinical outcomes has not been elucidated. The aim of this study was to assess whether timing of biologic initiation relative to CPI diagnosis impacts clinical and endoscopic remission.


Asunto(s)
Productos Biológicos , Colitis Ulcerosa , Reservorios Cólicos , Enfermedad de Crohn , Proctocolectomía Restauradora , Humanos , Colitis Ulcerosa/diagnóstico , Enfermedad de Crohn/diagnóstico , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/cirugía , Proctocolectomía Restauradora/efectos adversos , Inflamación/etiología
4.
Crit Care Med ; 47(5): e420-e427, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30730441

RESUMEN

OBJECTIVES: Sepsis is associated with a systemic inflammatory reaction, which can result in a life-endangering organ dysfunction. Pro-inflammatory responses during sepsis are characterized by increased activation of leukocytes and platelets, formation of platelet-neutrophil aggregates, and cytokine production. Sequestration of platelet-neutrophil aggregates in the microvasculature contributes to tissue damage during sepsis. At present no effective therapeutic strategy to ameliorate these events is available. In this preclinical pilot study, a novel anti-inflammatory approach was evaluated, which targets nucleoside triphosphate hydrolase activity toward activated platelets via a recombinant fusion protein combining a single-chain antibody against activated glycoprotein IIb/IIIa and the extracellular domain of CD39 (targ-CD39). DESIGN: Experimental animal study and cell culture study. SETTING: University-based experimental laboratory. SUBJECTS: Human dermal microvascular endothelial cells 1, human platelets and neutrophils, and C57BL/6NCrl mice. INTERVENTIONS: Platelet-leukocyte-endothelium interactions were evaluated under inflammatory conditions in vitro and in a murine lipopolysaccharide-induced sepsis model in vivo. The outcome of polymicrobial sepsis was evaluated in a murine cecal ligation and puncture model. To evaluate the anti-inflammatory potential of activated platelet targeted nucleoside triphosphate hydrolase activity, we employed a potato apyrase in vitro and in vivo, as well as targ-CD39 and as a control, nontarg-CD39 in vivo. MEASUREMENTS AND MAIN RESULTS: Under conditions of sepsis, agents with nucleoside triphosphate hydrolase activity decreased platelet-leukocyte-endothelium interaction, transcription of pro-inflammatory cytokines, microvascular platelet-neutrophil aggregate sequestration, activation marker expression on platelets and neutrophils contained in these aggregates, leukocyte extravasation, and organ damage. Targ-CD39 had the strongest effect on these variables and retained hemostasis in contrast to nontarg-CD39 and potato apyrase. Most importantly, targ-CD39 improved survival in the cecal ligation and puncture model to a stronger extent then nontarg-CD39 and potato apyrase. CONCLUSIONS: Targeting nucleoside triphosphate hydrolase activity (CD39) toward activated platelets is a promising new treatment concept to decrease systemic inflammation and mortality of sepsis. This innovative therapeutic approach warrants further development toward clinical application.


Asunto(s)
Plaquetas/metabolismo , Células Endoteliales/metabolismo , Sepsis/inmunología , Adenosina Trifosfatasas/farmacología , Animales , Plaquetas/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Proyectos Piloto
5.
Basic Res Cardiol ; 113(2): 11, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29344719

RESUMEN

Ischemic preconditioning (IP) is a well-known strategy to protect organs against cell death following ischemia. The previous work has shown that vasodilator-stimulated phosphoprotein (VASP) is involved in cytoskeletal reorganization and that it holds significant importance for the extent of myocardial ischemia reperfusion injury. Yet, the role of VASP during myocardial IP is, to date, not known. We report here that VASP phosphorylation at serine157 and serine239 is induced during hypoxia in vitro and during IP in vivo. The preconditioning-induced VASP phosphorylation inactivates the GP IIb/IIIa integrin receptor on platelets, which results in the reduced formation of organ compromising platelet neutrophil complexes. Experiments in chimeric mice confirmed the importance of VASP phosphorylation during myocardial IP. When studying this in VASP-/- animals and in an isolated heart model, we were able to confirm the important role of VASP on myocardial IP. In conclusion, we were able to show that IP-induced VASP phosphorylation in platelets is a protective mechanism against the deleterious effects of ischemia.


Asunto(s)
Plaquetas/metabolismo , Moléculas de Adhesión Celular/sangre , Precondicionamiento Isquémico Miocárdico/métodos , Proteínas de Microfilamentos/sangre , Infarto del Miocardio/prevención & control , Miocardio/metabolismo , Neutrófilos/metabolismo , Fosfoproteínas/sangre , Adhesividad Plaquetaria , Animales , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Hipoxia de la Célula , Modelos Animales de Enfermedad , Preparación de Corazón Aislado , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Infarto del Miocardio/sangre , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosforilación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 112(20): 6491-6, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25944935

RESUMEN

Platelets are crucial for hemostasis and thrombosis and exacerbate tissue injury following ischemia and reperfusion. Important regulators of platelet function are G proteins controlled by seven transmembrane receptors. The Gi protein Gα(i2) mediates platelet activation in vitro, but its in vivo role in hemostasis, arterial thrombosis, and postischemic infarct progression remains to be determined. Here we show that mice lacking Gα(i2) exhibit prolonged tail-bleeding times and markedly impaired thrombus formation and stability in different models of arterial thrombosis. We thus generated mice selectively lacking Gα(i2) in megakaryocytes and platelets (Gna(i2)(fl/fl)/PF4-Cre mice) and found bleeding defects comparable to those in global Gα(i2)-deficient mice. To examine the impact of platelet Gα(i2) in postischemic thrombo-inflammatory infarct progression, Gna(i2)(fl/fl)/PF4-Cre mice were subjected to experimental models of cerebral and myocardial ischemia/reperfusion injury. In the model of transient middle cerebral artery occlusion stroke Gna(i2)(fl/fl)/PF4-Cre mice developed significantly smaller brain infarcts and fewer neurological deficits than littermate controls. Following myocardial ischemia, Gna(i2)(fl/fl)/PF4-Cre mice showed dramatically reduced reperfusion injury which correlated with diminished formation of the ADP-dependent platelet neutrophil complex. In conclusion, our data provide definitive evidence that platelet Gα(i2) not only controls hemostatic and thrombotic responses but also is critical for the development of ischemia/reperfusion injury in vivo.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Inflamación/fisiopatología , Activación Plaquetaria/fisiología , Daño por Reperfusión/fisiopatología , Trombosis/fisiopatología , Animales , Tiempo de Sangría , Plaquetas/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/deficiencia , Immunoblotting , Megacariocitos/metabolismo , Ratones , Daño por Reperfusión/prevención & control
7.
Hepatology ; 63(5): 1689-705, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26573873

RESUMEN

UNLABELLED: Hepatic ischemia/reperfusion (I/R) is a major adverse reaction to liver transplantation, hemorrhagic shock, or resection. Recently, the anti-inflammatory properties of the axonal guidance cue netrin-1 were reported. Here, we demonstrate that netrin-1 also impacts the resolution of inflammation and promotes hepatic repair and regeneration during liver I/R injury. In initial studies, we investigated the induction of netrin-1 and its receptors in murine liver tissues after I/R injury. Hepatic I/R injury was performed in mice with a partial genetic netrin-1 deficiency (Ntn1(+/-) ) or wild-type C57BL/6 treated with exogenous netrin-1 to examine the endogenous and therapeutically administered impact of netrin-1. These investigations were corroborated by studies determining the characteristics of intravascular leukocyte flow, clearance of apoptotic neutrophils (polymorphonuclear cells [PMNs]), production of specialized proresolving lipid mediators (SPMs), generation of specific growth factors contributing to the resolution of inflammation, and liver repair. Hepatic I/R was associated with a significant reduction of netrin-1 transcript and protein in murine liver tissue. Subsequent studies in netrin-1-deficient mice revealed lower efficacies in reducing PMN infiltration, proinflammatory cytokine levels, and hepatic-specific injury enzymes. Conversely, mice treated with exogenous netrin-1 exhibited increased liver protection and repair, reducing neutrophil influx into the injury site, decreasing proinflammatory mediators, increasing efferocytosis of apoptotic PMNs, and stimulating local endogenous biosynthesis of SPMs and the generation of specific growth factors. Finally, genetic studies implicated the A2B adenosine receptor in netrin-1-mediated protection during hepatic I/R injury. CONCLUSION: The present study indicates a previously unrecognized role for netrin-1 in liver protection and its contribution to tissue homeostasis and regeneration.


Asunto(s)
Regeneración Hepática , Factores de Crecimiento Nervioso/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Hepatitis/fisiopatología , Humanos , Lipoxinas/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Netrina , Netrina-1 , Neutrófilos/fisiología , Receptores de Superficie Celular/fisiología , Daño por Reperfusión/fisiopatología
8.
Crit Care Med ; 44(8): e625-32, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26963322

RESUMEN

OBJECTIVES: Hepatic ischemia-reperfusion injury is a disease pattern that is associated with an acute inflammatory reaction. It is well known that neutrophils play an essential role in the early phase of hepatic ischemia-reperfusion injury and determine the extent of tissue damage. Hepatic ischemia-reperfusion injury can result in organ failure, which is linked to high mortality. Recent data indicate that the neuronal guidance receptor Plexin C1 is involved in the control of the acute inflammatory response and, as such, modulates the transmigration of neutrophils. Hence, we investigated the functional role of Plexin C1 in a mouse model of early hepatic ischemia-reperfusion injury. DESIGN: Animal study. SETTING: University experimental laboratory. SUBJECTS: Wild-type, PLXNC1 and chimeric mice. INTERVENTIONS: Hepatic ischemia-reperfusion injury or sham operation. MEASUREMENTS AND MAIN RESULTS: We found that the functional inhibition of Plexin C1 in wild-type mice treated with an anti-Plexin C1 antibody and a Semaphorin 7A peptide reduced hepatic ischemia-reperfusion injury, as measured by the levels of lactate dehydrogenase, aspartate, and alanine aminotransferase. This reduction in ischemia-reperfusion injury was accompanied by reduced numbers of neutrophils in ischemic hepatic tissue and reduced serum levels of inflammatory cytokines. Experiments using Plexin C1 receptor-deficient (PLXNC1) mice also demonstrated decreased hepatic ischemia-reperfusion injury. Studies of chimeric mice revealed that the hematopoietic Plexin C1 knockout is crucial for reducing the extent of hepatic ischemia-reperfusion injury. CONCLUSIONS: These results describe a role for Plexin C1 during ischemia-reperfusion injury, highlight the role of hematopoietic Plexin C1 in the development of hepatic ischemia-reperfusion injury, and suggest that Plexin C1 is a potential drug target.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/agonistas , Hígado/fisiopatología , Daño por Reperfusión/prevención & control , Daño por Reperfusión/fisiopatología , Alanina Transaminasa/metabolismo , Animales , Antígenos CD/farmacología , Ácido Aspártico/metabolismo , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/farmacología , Mediadores de Inflamación/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Neutrófilos/metabolismo , Semaforinas/farmacología
9.
Crit Care Med ; 44(4): e181-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26491864

RESUMEN

OBJECTIVES: Extracellular adenosine has tissue-protective potential in several conditions. Adenosine levels are regulated by a close interplay between nucleoside transporters and adenosine kinase. On the basis of the evidence of the role of adenosine kinase in regulating adenosine levels during hypoxia, we evaluated the effect of adenosine kinase on lung injury. Furthermore, we tested the influence of a pharmacologic approach to blocking adenosine kinase on the extent of lung injury. DESIGN: Prospective experimental animal study. SETTING: University-based research laboratory. SUBJECTS: In vitro cell lines, wild-type and adenosine kinase+/- mice. INTERVENTIONS: We tested the expression of adenosine kinase during inflammatory stimulation in vitro and in a model of lipopolysaccharide inhalation in vivo. Studies using the adenosine kinase promoter were performed in vitro. Wild-type and adenosine kinase+/- mice were subjected to lipopolysaccharide inhalation. Pharmacologic inhibition of adenosine kinase was performed in vitro, and its effect on adenosine uptake was evaluated. The pharmacologic inhibition was also performed in vivo, and the effect on lung injury was assessed. MEASUREMENTS AND MAIN RESULTS: We observed the repression of adenosine kinase by proinflammatory cytokines and found a significant influence of nuclear factor kappa-light-chain-enhancer of activated B-cells on regulation of the adenosine kinase promoter. Mice with endogenous adenosine kinase repression (adenosine kinase+/-) showed reduced infiltration of leukocytes into the alveolar space, decreased total protein and myeloperoxidase levels, and lower cytokine levels in the alveolar lavage fluid. The inhibition of adenosine kinase by 5-iodotubercidin increased the extracellular adenosine levels in vitro, diminished the transmigration of neutrophils, and improved the epithelial barrier function. The inhibition of adenosine kinase in vivo showed protective properties, reducing the extent of pulmonary inflammation during lung injury. CONCLUSIONS: Taken together, these data show that adenosine kinase is a valuable target for reducing the inflammatory changes associated with lung injury and should be pursued as a therapeutic option.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Adenosina Quinasa/antagonistas & inhibidores , Pulmón/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Linfocitos B/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Línea Celular , Citocinas/metabolismo , Lipopolisacáridos/administración & dosificación , Ratones , Neumonía/metabolismo , Estudios Prospectivos , Tubercidina/análogos & derivados , Tubercidina/farmacología
10.
Anesthesiology ; 125(3): 547-60, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27404219

RESUMEN

BACKGROUND: Liver ischemia/reperfusion (IR) injury is characterized by hepatic tissue damage and an inflammatory response. This is accompanied by the formation and vascular sequestration of platelet-neutrophil conjugates (PNCs). Signaling through Adora2b adenosine receptors can provide liver protection. Volatile anesthetics may interact with adenosine receptors. This study investigates potential antiinflammatory effects of the volatile anesthetic sevoflurane during liver IR. METHODS: Experiments were performed ex vivo with human blood and in a liver IR model with wild-type, Adora2a, and Adora2b mice. The effect of sevoflurane on platelet activation, PNC formation and sequestration, cytokine release, and liver damage (alanine aminotransferase release) was analyzed using flow cytometry, luminometry, and immunofluorescence. Adenosine receptor expression in liver tissue was analyzed using immunohistochemistry and real-time polymerase chain reaction. RESULTS: Ex vivo experiments indicate that sevoflurane inhibits platelet and leukocyte activation (n = 5). During liver IR, sevoflurane (2 Vol%) decreased PNC formation 2.4-fold in wild-type (P < 0.05) but not in Adora2b mice (n ≥ 5). Sevoflurane reduced PNC sequestration 1.9-fold (P < 0.05) and alanine aminotransferase release 3.5-fold (P < 0.05) in wild-type but not in Adora2b mice (n = 5). In Adora2a mice, sevoflurane also inhibited PNC formation and cytokine release. Sevoflurane diminished cytokine release (n ≥ 3) and increased Adora2b transcription and expression in liver tissue of wild-types (n = 4). CONCLUSIONS: Our experiments highlight antiinflammatory and tissue-protective properties of sevoflurane during liver IR and reveal a mechanistic role of Adora2b in sevoflurane-associated effects. The targeted use of sevoflurane not only as an anesthetic but also to prevent IR damage is a promising approach in the treatment of critically ill patients.


Asunto(s)
Anestésicos por Inhalación/farmacología , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , Éteres Metílicos/farmacología , Receptor de Adenosina A2B/metabolismo , Daño por Reperfusión/prevención & control , Adulto , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Adenosina A2B/genética , Sevoflurano , Transducción de Señal
11.
Eur J Immunol ; 44(9): 2648-58, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24890788

RESUMEN

Acute inflammation is the pathophysiological basis of important clinical conditions associated with organ failure. The initial inflammatory response is controlled by the chemokine system, yet recent data have indicated that the neuronal guidance cues are significantly involved in the orchestration of this process. Previous work has shown the proinflammatory capacity of the guidance cue semaphorin (Sema) 7a, but the role of one of its target receptors, the plexin C1 (PLXNC1) receptor is to date unknown. We report here that PLXNC1 is expressed outside the nervous system and induced during acute inflammation. PLXNC1(-/-) mice with C57BL/6 background demonstrated decreased inflammatory responses during zymosan A (ZyA)-induced peritonitis. Subsequent in vivo studies revealed altered rolling, adhesion, and transmigration properties of PLXNC1(-/-) leukocytes. Blockade of PLXNC1 was associated with attenuated chemotactic transendothelial migration properties in vitro. Studies in chimeric mice revealed that hematopoietic PLXNC1(-/-) animals demonstrated an attenuated inflammatory response. To probe the therapeutic potential of PLXNC1 we treated C57BL/6 WT mice with an anti-PLXNC1 antibody and a PLXNC1 binding peptide. Both of these interventions significantly dampened ZyA-induced peritonitis. These results implicate an important role of PLXNC1 during an acute inflammatory response and indicate PLXNC1 as a potential target for the control of conditions associated with acute inflammation.


Asunto(s)
Proteínas del Tejido Nervioso/inmunología , Peritonitis/inmunología , Receptores de Superficie Celular/inmunología , Enfermedad Aguda , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Peritonitis/inducido químicamente , Peritonitis/genética , Peritonitis/patología , Receptores de Superficie Celular/genética , Semaforinas/genética , Semaforinas/inmunología , Zimosan/toxicidad
12.
Proc Natl Acad Sci U S A ; 109(35): 14146-51, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891341

RESUMEN

Recent studies identified basic biological principles that are shared by the immune and the nervous system. One of these analogies applies to the orchestration of cellular migration where guidance proteins that serve as a stop signal for axonal migration can also serve as a stop signal for the migration of immune-competent cells. The control of leukocyte migration is of key interest during conditions associated with inflammatory tissue changes such as tissue hypoxia or hypoxic inflammation. Semaphorins are members of these axon guidance molecules. Previously unknown, we report here the expression and induction of semaphorin 7A (SEMA7A) on endothelium through hypoxia-inducible factor 1α during hypoxia. This induction of SEMA7A translates into increased transmigration of polymorphonuclear neutrophil granulocytes across endothelial cells. Extension of these findings demonstrated an attenuated extravasation of polymorphonuclear neutrophil granulocytes in Sema7a-deficient mice from the vasculature during hypoxia. Studies using chimeric animals identified the expression of Sema7A on nonhematopoietic tissue to be the underlying cause of the observed results. Taken together, our findings demonstrate that neuronal guidance proteins do not only serve as a stop signal for leukocyte migration but also can propagate the extravasation of leukocytes from the vascular space. Future anti-inflammatory strategies might be based on this finding.


Asunto(s)
Antígenos CD/inmunología , Movimiento Celular/inmunología , Hipoxia/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Semaforinas/inmunología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Trasplante de Médula Ósea , Endotelio/citología , Endotelio/inmunología , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Neutrófilos/citología , ARN Interferente Pequeño/genética , Semaforinas/genética , Semaforinas/metabolismo
13.
Proc Natl Acad Sci U S A ; 108(16): 6555-60, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21467223

RESUMEN

Directed cell migration is a prerequisite not only for the development of the central nervous system, but also for topically restricted, appropriate immune responses. This is crucial for host defense and immune surveillance. Attracting environmental cues guiding leukocyte cell traffic are likely to be complemented by repulsive cues, which actively abolish cell migration. One such a paradigm exists in the developing nervous system, where neuronal migration and axonal path finding is balanced by chemoattractive and chemorepulsive cues, such as the neuronal repulsive guidance molecule-A (RGM-A). As expressed at the inflammatory site, the role of RGM-A within the immune response remains unclear. Here we report that RGM-A (i) is expressed by epithelium and leukocytes (granulocytes, monocytes, and T/B lymphocytes); (ii) inhibits leukocyte migration by contact repulsion and chemorepulsion, depending on dosage, through its receptor neogenin; and (iii) suppresses the inflammatory response in a model of zymosan-A-induced peritonitis. Systemic application of RGM-A attenuates the humoral proinflammatory response (TNF-α, IL-6, and macrophage inflammatory protein 1α), infiltration of inflammatory cell traffic, and edema formation. In contrast, the demonstrated anti-inflammatory effect of RGM-A is absent in mice homozygous for a gene trap mutation in the neo1 locus (encoding neogenin). Thus, our results suggest that RGM-A is a unique endogenous inhibitor of leukocyte chemotaxis that limits inflammatory leukocyte traffic and creates opportunities to better understand and treat pathologies caused by exacerbated or misdirected inflammatory responses.


Asunto(s)
Quimiotaxis/inmunología , Regulación de la Expresión Génica/inmunología , Leucocitos/inmunología , Proteínas del Tejido Nervioso/inmunología , Peritonitis/inmunología , Animales , Células CACO-2 , Quimiotaxis/efectos de los fármacos , Quimiotaxis/genética , Citocinas/biosíntesis , Citocinas/genética , Citocinas/inmunología , Epitelio/inmunología , Epitelio/metabolismo , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Leucocitos/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Peritonitis/inducido químicamente , Peritonitis/genética , Peritonitis/metabolismo , Zimosan/toxicidad
14.
Nutr Clin Pract ; 2024 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-38922984

RESUMEN

Malnutrition is estimated to affect roughly 30%-80% of patients with inflammatory bowel disease (IBD). In those patients who cannot tolerate sufficient oral nutrition or there is no possibility for placing an enteral nutrition tube, parenteral nutrition offers a lifesaving alternative. However, this is not without risk. For patients with IBD, understanding the indications, contraindications, and complications associated with parenteral nutrition is crucial. In this review, we will discuss the indications and contraindications for parenteral nutrition in patients with IBD, the common complications associated with intravenous nutrition, the use of parenteral nutrition in special populations, such as in pediatric and perioperative patients, and the impact of parenteral nutrition on IBD-related outcomes.

15.
Blood Adv ; 8(11): 2660-2674, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38489236

RESUMEN

ABSTRACT: Pulmonary defense mechanisms are critical for host integrity during pneumonia and sepsis. This defense is fundamentally dependent on the activation of neutrophils during the innate immune response. Recent work has shown that semaphorin 7A (Sema7A) holds significant impact on platelet function, yet its role on neutrophil function within the lung is not well understood. This study aimed to identify the role of Sema7A during pulmonary inflammation and sepsis. In patients with acute respiratory distress syndrome (ARDS), we were able to show a correlation between Sema7A and oxygenation levels. During subsequent workup, we found that Sema7A binds to the neutrophil PlexinC1 receptor, increasing integrins, and L-selectin on neutrophils. Sema7A prompted neutrophil chemotaxis in vitro and the formation of platelet-neutrophil complexes in vivo. We also observed altered adhesion and transmigration of neutrophils in Sema7A-/-animals in the lung during pulmonary inflammation. This effect resulted in increased number of neutrophils in the interstitial space of Sema7A-/- animals but reduced numbers of neutrophils in the alveolar space during pulmonary sepsis. This finding was associated with significantly worse outcome of Sema7A-/- animals in a model of pulmonary sepsis. Sema7A has an immunomodulatory effect in the lung, affecting pulmonary sepsis and ARDS. This effect influences the response of neutrophils to external aggression and might influence patient outcome. This trial was registered at www.ClinicalTrials.gov as #NCT02692118.


Asunto(s)
Antígenos CD , Neutrófilos , Neumonía , Semaforinas , Sepsis , Semaforinas/metabolismo , Sepsis/inmunología , Sepsis/metabolismo , Neutrófilos/metabolismo , Neutrófilos/inmunología , Humanos , Animales , Ratones , Antígenos CD/metabolismo , Neumonía/metabolismo , Neumonía/inmunología , Proteínas Ligadas a GPI/metabolismo , Masculino , Modelos Animales de Enfermedad , Ratones Noqueados , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/metabolismo , Femenino
16.
Am J Respir Cell Mol Biol ; 49(2): 296-305, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23590299

RESUMEN

Acute lung injury (ALI) is a devastating disorder of the lung that is characterized by hypoxemia, overwhelming pulmonary inflammation, and a high mortality in the critically ill. Adenosine has been implicated as an anti-inflammatory signaling molecule, and previous studies showed that extracellular adenosine concentrations are increased in inflamed tissues. Adenosine signaling is terminated by the uptake of adenosine from the extracellular into the intracellular compartment via equilibrative nucleoside transporters (ENTs). However, their role in controlling adenosine signaling during pulmonary inflammation remains unknown. After inflammatory in vitro experiments, we observed a repression of ENT1 and ENT2 that was associated with an attenuation of extracellular adenosine uptake. Experiments using short, interfering RNA silencing confirmed a significant contribution of ENT repression in elevating extracellular adenosine concentrations during inflammation. Furthermore, an examination of the ENT2 promoter implicated NF-κB as a key regulator for the observed ENT repression. Additional in vivo experiments using a murine model of inflammatory lung injury showed that the pharmacological inhibition of ENT1 and ENT2 resulted in improved pulmonary barrier function and reduced signs of acute inflammation of the lung. Whereas experiments on Ent1(-/-) or Ent2(-/-) mice revealed lung protection in LPS-induced lung injury, an examination of bone marrow chimeras for ENTs pointed to the nonhematopoetic expression of ENTs as the underlying cause of dampened pulmonary inflammation during ALI. Taken together, these findings reveal the transcriptional repression of ENTs as an innate protective response during acute pulmonary inflammation. The inhibition of ENTs could be pursued as a therapeutic option to ameliorate inflammatory lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/biosíntesis , Transportador Equilibrativo 2 de Nucleósido/biosíntesis , Pulmón/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Línea Celular , Tranportador Equilibrativo 1 de Nucleósido/genética , Transportador Equilibrativo 2 de Nucleósido/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Pulmón/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal/genética
17.
Microsurgery ; 33(3): 216-22, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23180484

RESUMEN

INTRODUCTION: In this study, we evaluated the role of the Netrin-1 receptor UNC5b (Uncoordinated), a neuronal guidance molecule, during peripheral nerve regeneration using the mouse median nerve model. MATERIALS AND METHODS: Using Western blot analysis, we examined the expression changes of UNC5b after transection and microsurgical repair of the mouse median nerve distal to the transection site. We evaluated the histomorphometrical changes and functional recovery of the grasping force after median nerve transection and repair in wild-type (WT) mice and UNC5b(+/-) heterozygous mice. RESULTS: In Western blot analysis, we could show a high increase of UNC5b in the nerve segment distal to the injury site at day 14. Histomorphometrical analysis did not show any significant differences between WT animals and heterozygous animals. Using the functional grasping test, we could demonstrate that peripheral nerve regeneration is significantly diminished in heterozygous UNC5b(+/-) mice. CONCLUSION: By using the mouse median nerve model in transgenic animals, we demonstrate that the Netrin-1 receptor UNC5b plays an important role during peripheral nerve regeneration.


Asunto(s)
Regeneración Nerviosa/fisiología , Receptores de Superficie Celular/biosíntesis , Animales , Nervio Mediano/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Receptores de Netrina , Receptores de Superficie Celular/fisiología , Factores de Tiempo
18.
Front Immunol ; 14: 1251026, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38094294

RESUMEN

Introduction: The study explores the role of endothelial Semaphorin 7A (SEMA7A) in inflammatory processes. SEMA7A is known for enhancing inflammation during tissue hypoxia and exhibiting anti-inflammatory properties in the intestinal system during colitis. This research extends the understanding of SEMA7A's function by examining its role in inflammatory peritonitis and intestinal inflammation. Methods: The research involved inducing peritonitis in SEMA7A knockout (SEMA7A-/-) and wild-type (WT) animals through Zymosan A (ZyA) injection. The inflammatory response was assessed by measuring cell count and cytokine release. In parallel, the study investigated the expression of SEMA7A in intestinal epithelial cells under inflammatory stimuli and its impact on interleukin 10 (IL-10) production using an in vitro co-culture model of monocytes and epithelial cells. Additionally, the distribution of SEMA7A target receptors, particularly ITGAV/ITGB1 (CD51/CD29), was analyzed in WT animals. Results: The results revealed that SEMA7A-/- animals exhibited increased inflammatory peritonitis compared to the WT animals. Inflammatory conditions in intestinal epithelial cells led to the induction of SEMA7A. The co-culture experiments demonstrated that SEMA7A induced IL-10 production, which depended on integrin receptors and was independent of PLXNC1 expression. Furthermore, ITGAV/ITGB1 emerged as the predominant SEMA7A receptor in the intestinal area of WT animals. Discussion: These findings underscore the multifaceted role of SEMA7A in inflammatory processes. The differential responses in peritonitis and intestinal inflammation suggest that SEMA7A's function is significantly influenced by the expression and distribution of its target receptors within different organ systems. The study highlights the complex and context-dependent nature of SEMA7A in mediating inflammatory responses.


Asunto(s)
Peritonitis , Semaforinas , Animales , Antígenos CD/metabolismo , Integrinas , Interleucina-10/genética , Semaforinas/genética , Semaforinas/metabolismo , Peritonitis/inducido químicamente , Inflamación
19.
Remote Sens (Basel) ; 15(19): 1-25, 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-38362160

RESUMEN

Mapping the seagrass distribution and density in the underwater landscape can improve global Blue Carbon estimates. However, atmospheric absorption and scattering introduce errors in space-based sensors' retrieval of sea surface reflectance, affecting seagrass presence, density, and above-ground carbon (AGCseagrass) estimates. This study assessed atmospheric correction's impact on mapping seagrass using WorldView-2 satellite imagery from Saint Joseph Bay, Saint George Sound, and Keaton Beach in Florida, USA. Coincident in situ measurements of water-leaving radiance (LW), optical properties, and seagrass leaf area index (LAI) were collected. Seagrass classification and the retrieval of LAI were compared after empirical line height (ELH) and dark-object subtraction (DOS) methods were used for atmospheric correction. DOS left residual brightness in the blue and green bands but had minimal impact on the seagrass classification accuracy. However, the brighter reflectance values reduced LAI retrievals by up to 50% compared to ELH-corrected images and ground-based observations. This study offers a potential correction for LAI underestimation due to incomplete atmospheric correction, enhancing the retrieval of seagrass density and above-ground Blue Carbon from WorldView-2 imagery without in situ observations for accurate atmospheric interference correction.

20.
Circulation ; 123(22): 2579-90, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21606399

RESUMEN

BACKGROUND: Recent work has suggested that the formation of platelet-neutrophil complexes (PNCs) aggravates the severity of inflammatory tissue injury. Given the importance of vasodilator-stimulated phosphoprotein (VASP) for platelet function, we pursued the role of VASP on the formation of PNCs and its impact on the extent of myocardial ischemia-reperfusion (IR) injury. METHODS AND RESULTS: In initial in vitro studies we found that neutrophils facilitated the movement of platelets across endothelial monolayers. Phosphorylation of VASP reduced the formation of PNCs and transendothelial movement of PNCs. During myocardial IR injury, VASP(-/-) animals demonstrated reduced intravascular formation of PNCs and reduced presence of PNCs within the ischemic myocardial tissue. This was associated with reduced IR injury. Studies using platelet transfer and bone marrow chimeric animals showed that hematopoietic VASP expression was crucial for the intravascular formation of PNCs the presence of PNCs within ischemic myocardial tissue and the extent of myocardial IR injury. Furthermore, phosphorylation of VASP on Ser153 or Ser235 reduced intravascular PNC formation and presence of PNCs within ischemic myocardial tissue. This finding was associated with reduced myocardial IR injury. CONCLUSION: Previously unappreciated, the phosphorylation of VASP performs a key function for the formation of PNCs that is crucially important for the extent of myocardial IR injury.


Asunto(s)
Plaquetas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de Microfilamentos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Neutrófilos/metabolismo , Fosfoproteínas/metabolismo , Animales , Plaquetas/citología , Movimiento Celular/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Fosforilación/fisiología , Quimera por Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA