Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Front Immunol ; 14: 1158045, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37090721

RESUMEN

Colorectal cancer (CRC) is the third most predominant malignancy in the world. Although the importance of immune system in cancer development has been well established, the underlying mechanisms remain to be investigated further. Here we studied a novel protein prokineticin 2 (Prok2, also known as Bv8) as a key pro-tumoral factor in CRC progression in in vitro and ex vivo settings. Human colorectal tumor tissues, myeloid cell lines (U937 cells and HL60 cells) and colorectal cancer cell line (Caco-2 cells) were used for various studies. Myeloid cell infiltration (especially neutrophils) and Bv8 accumulation were detected in human colorectal tumor tissue with immunostaining. The chemotactic effects of Bv8 on myeloid cells were presented in the transwell assay and chemotaxis assy. Cultured CRC cells treated with myeloid cells or Bv8 produced reactive oxygen species (ROS) and vascular endothelial growth factor (VEGF). Furthermore, ROS and VEGF acted as pro-angiogenesis buffer in myeloid cell-infiltrated CRC microenvironment. Moreover, myeloid cells or Bv8 enhanced energy consumption of glycolysis ATP and mitochondria ATP of CRC cells. Interestingly, myeloid cells increased CRC cell viability, but CRC cells decreased the viability of myeloid cells. ERK signalling pathway in CRC cells was activated in the presence of Bv8 or co-cultured myeloid cells. In conclusion, our data indicated the vital roles of Bv8 in myeloid cell infiltration and CRC development, suggesting that Bv8 may be a potential therapeutic target for colorectal cancer-related immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Neuropéptidos , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células CACO-2 , Especies Reactivas de Oxígeno/metabolismo , Neuropéptidos/metabolismo , Células Mieloides/metabolismo , Movimiento Celular , Neoplasias Colorrectales/patología , Factores de Crecimiento Endotelial Vascular/metabolismo , Adenosina Trifosfato/metabolismo , Microambiente Tumoral
2.
Front Immunol ; 13: 853769, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572508

RESUMEN

Intra-alveolar microvesicles (MVs) are important mediators of inter-cellular communication within the alveolar space, and are key components in the pathophysiology of lung inflammation such as acute respiratory distress syndrome (ARDS). Despite the abundance of data detailing the pro-inflammatory effects of MVs, it remains unclear how MVs interact or signal with target cells in the alveolus. Using both in vivo and in vitro alveolar models, we analyzed the dynamics of MV uptake by resident alveolar cells: alveolar macrophages and epithelial cells. Under resting conditions, the overwhelming majority of MVs were taken up by alveolar macrophages. However, following lipopolysaccharide (LPS)-mediated inflammation, epithelial cells internalized significantly more MVs (p<0.01) whilst alveolar macrophage internalization was significantly reduced (p<0.01). We found that alveolar macrophages adopted a pro-inflammatory phenotype after internalizing MVs under resting conditions, but reduction of MV uptake following LPS pre-treatment was associated with loss of inflammatory phenotype. Instead, MVs induced significant epithelial cell inflammation following LPS pre-treatment, when MV internalization was most significant. Using pharmacological inhibitors, we interrogated the mechanisms of MV internalization to identify which endocytic pathways and cell surface receptors are involved. We demonstrated that epithelial cells are exclusively dependent on the clathrin and caveolin dependent endocytotic pathway, whereas alveolar macrophage uptake may involve a significant phagocytic component. Furthermore, alveolar macrophages predominantly engulf MVs via scavenger receptors whilst, epithelial cells internalize MVs via a phosphatidylserine/integrin receptor mediated pathway (specifically alpha V beta III), which can be inhibited with phosphatidylserine-binding protein (i.e. annexin V). In summary, we have undertaken a comprehensive evaluation of MV internalization within the alveolar space. Our results demonstrate that different environmental conditions can modulate MV internalization, with inflammatory stimuli strongly enhancing epithelial cell uptake of MVs and inducing epithelial cell activation. Our data reveal the unique mechanisms by which alveolar macrophages and epithelial cells internalize MVs thereby elucidating how MVs exert their pathophysiological effect during lung inflammation and injury. As MVs are potential novel therapeutic targets in conditions such as ARDS, these data provide crucial insights into the dynamics of MV-target cell interactions and highlight potential avenues for researchers to modulate and inhibit their pro-inflammatory actions within the alveolar space.


Asunto(s)
Neumonía , Síndrome de Dificultad Respiratoria , Células Epiteliales , Humanos , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos Alveolares/metabolismo , Fosfatidilserinas/metabolismo , Neumonía/metabolismo
3.
Shock ; 53(2): 199-207, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30998647

RESUMEN

Our previous work has shown that pregnancy exacerbates the hypotensive response to both infection and lipopolysaccharide (LPS). The high levels of progesterone (P4) associated with pregnancy have been suggested to be responsible for the pregnancy-induced changes in the cardiovascular response to infection. Here, we test the hypothesis that P4 supplementation exacerbates the hypotensive response of the maternal cardiovascular to LPS.Female CD1 mice had radiotelemetry probes implanted to measure hemodynamic function noninvasively and were time-mated. From day 14 of pregnancy, mice received either 10 mg of P4 or vehicle alone per day and on day 16, intraperitoneal LPS (10 µg of serotype 0111:B4) was injected. In two identically treated cohorts of mice, tissue and serum (for RNA, protein studies) were collected at 6 and 12 h.Administration of LPS resulted in a fall in blood pressure in vehicle treated, but not P4 supplemented mice. This occurred with similar changes in the circulating levels of cytokines, vasoactive factors and in both circulating and tissue inflammatory cell numbers, but with reduced left ventricular expression of cytokines in P4-supplemented mice. However, left ventricular expression of markers of cardiac dysfunction and apoptosis were similar.This study demonstrates that P4 supplementation prevented LPS-induced hypotension in pregnant mice in association with reduced myocardial inflammatory cytokine gene expression. These observations suggest that rather than being detrimental, P4 supplementation has a protective effect on the maternal cardiovascular response to sepsis.


Asunto(s)
Hipotensión/inducido químicamente , Hipotensión/tratamiento farmacológico , Lipopolisacáridos/toxicidad , Progesterona/uso terapéutico , Animales , Biomarcadores/metabolismo , Presión Sanguínea/efectos de los fármacos , Femenino , Citometría de Flujo , Ratones , Embarazo
4.
Biol Reprod ; 102(2): 445-455, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31599921

RESUMEN

In our earlier work, we found that intrauterine (i.u.) and intraperitoneal (i.p.) injection of LPS (10-µg serotype 0111:B4) induced preterm labor (PTL) with high pup mortality, marked systemic inflammatory response and hypotension. Here, we used both i.u. and i.p. LPS models in pregnant wild-type (wt) and CCR2 knockout (CCR2-/-) mice on E16 to investigate the role played by the CCL2/CCR2 system in the response to LPS. Basally, lower numbers of monocytes and macrophages and higher numbers of neutrophils were found in the myometrium, placenta, and blood of CCR2-/- vs. wt mice. After i.u. LPS, parturition occurred at 14 h in both groups of mice. At 7 h post-injection, 70% of wt pups were dead vs. 10% of CCR2-/- pups, but at delivery 100% of wt and 90% of CCR2-/- pups were dead. Myometrial and placental monocytes and macrophages were generally lower in CCR2-/- mice, but this was less consistent in the circulation, lung, and liver. At 7 h post-LPS, myometrial ERK activation was greater and JNK and p65 lower and the mRNA levels of chemokines were higher and of inflammatory cytokines lower in CCR2-/- vs. wt mice. Pup brain and placental inflammation were similar. Using the IP LPS model, we found that all measures of arterial pressure increased in CCR2-/- but declined in wt mice. These data suggest that the CCL2/CCR2 system plays a critical role in the cardiovascular response to LPS and contributes to pup death but does not influence the onset of inflammation-induced PTL.


Asunto(s)
Presión Arterial/fisiología , Lipopolisacáridos/efectos adversos , Miometrio/metabolismo , Trabajo de Parto Prematuro/inducido químicamente , Placenta/metabolismo , Receptores CCR2/metabolismo , Animales , Presión Arterial/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Miometrio/efectos de los fármacos , Trabajo de Parto Prematuro/genética , Trabajo de Parto Prematuro/metabolismo , Parto/efectos de los fármacos , Parto/genética , Parto/metabolismo , Placenta/efectos de los fármacos , Embarazo , Receptores CCR2/genética
5.
FASEB J ; 33(5): 6442-6455, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30776316

RESUMEN

Cellular stress or injury induces release of endogenous danger signals such as ATP, which plays a central role in activating immune cells. ATP is essential for the release of nonclassically secreted cytokines such as IL-1ß but, paradoxically, has been reported to inhibit the release of classically secreted cytokines such as TNF. Here, we reveal that ATP does switch off soluble TNF (17 kDa) release from LPS-treated macrophages, but rather than inhibiting the entire TNF secretion, ATP packages membrane TNF (26 kDa) within microvesicles (MVs). Secretion of membrane TNF within MVs bypasses the conventional endoplasmic reticulum- and Golgi transport-dependent pathway and is mediated by acid sphingomyelinase. These membrane TNF-carrying MVs are biologically more potent than soluble TNF in vivo, producing significant lung inflammation in mice. Thus, ATP critically alters TNF trafficking and secretion from macrophages, inducing novel unconventional membrane TNF signaling via MVs without direct cell-to-cell contact. These data have crucial implications for this key cytokine, particularly when therapeutically targeting TNF in acute inflammatory diseases.-Soni, S., O'Dea, K. P., Tan, Y. Y., Cho, K., Abe, E., Romano, R., Cui, J., Ma, D., Sarathchandra, P., Wilson, M. R., Takata, M. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles.


Asunto(s)
Adenosina Trifosfato/inmunología , Membrana Celular/inmunología , Vesículas Extracelulares/inmunología , Macrófagos/inmunología , Neumonía/inmunología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Enfermedad Aguda , Adenosina Trifosfato/genética , Animales , Comunicación Celular/genética , Comunicación Celular/inmunología , Membrana Celular/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Vesículas Extracelulares/genética , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/genética
6.
Sci Rep ; 8(1): 14736, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30283005

RESUMEN

Clinical investigations lack predictive value when diagnosing pneumonia, especially when patients are ventilated and develop ventilator associated pneumonia (VAP). New tools to aid diagnosis are important to improve outcomes. This pilot study examines the potential for a panel of inflammatory mediators to aid in the diagnosis. Forty-four ventilated patients, 17 with pneumonia and 27 with brain injuries, eight of whom developed VAP, were recruited. 51 inflammatory mediators, including cytokines and oxylipins, were measured in patients' serum using flow cytometry and mass spectrometry. The mediators could separate patients admitted to ICU with pneumonia compared to brain injury with an area under the receiver operating characteristic curve (AUROC) 0.75 (0.61-0.90). Changes in inflammatory mediators were similar in both groups over the course of ICU stay with 5,6-dihydroxyeicosatrienoic and 8,9-dihydroxyeicosatrienoic acids increasing over time and interleukin-6 decreasing. However, brain injured patients who developed VAP maintained inflammatory profiles similar to those at admission. A multivariate model containing 5,6-dihydroxyeicosatrienoic acid, 8,9-dihydroxyeicosatrienoic acid, intercellular adhesion molecule-1, interleukin-6, and interleukin-8, could differentiate patients with VAP from brain injured patients without infection (AUROC 0.94 (0.80-1.00)). The use of a selected group of markers showed promise to aid the diagnosis of VAP especially when combined with clinical data.


Asunto(s)
Biomarcadores/sangre , Lesiones Encefálicas/sangre , Inflamación/sangre , Neumonía Asociada al Ventilador/sangre , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Cuidados Críticos , Femenino , Citometría de Flujo , Humanos , Inflamación/fisiopatología , Unidades de Cuidados Intensivos , Interleucina-6/sangre , Interleucina-8/sangre , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Neumonía Asociada al Ventilador/fisiopatología , Curva ROC
7.
Biol Reprod ; 98(3): 376-395, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29145579

RESUMEN

The role of progesterone (P4) in the regulation of the local (uterine) and systemic innate immune system, myometrial expression of connexin 43 (Cx-43) and cyclooxygenase 2 (COX-2), and the onset of parturition was examined in (i) naïve mice delivering at term; (ii) E16 mice treated with RU486 (P4-antagonist) to induce preterm parturition; and (iii) in mice treated with P4 to prevent term parturition. In naïve mice, myometrial neutrophil and monocyte numbers peaked at E18 and declined with the onset of parturition. In contrast, circulating monocytes did not change and although neutrophils were increased with pregnancy, they did not change across gestation. The myometrial mRNA and protein levels of most chemokines/cytokines, Cx-43, and COX-2 increased with, but not before, parturition. With RU486-induced parturition, myometrial and systemic neutrophil numbers increased before and myometrial monocyte numbers increased with parturition only. Myometrial chemokine/cytokine mRNA abundance increased with parturition, but protein levels peaked earlier at between 4.5 and 9 h post-RU486. Cx-43, but not COX-2, mRNA expression and protein levels increased prior to the onset of parturition. In mice treated with P4, the gestation-linked increase in myometrial monocyte, but not neutrophil, numbers was prevented, and expression of Cx-43 and COX-2 was reduced. On E20 of P4 supplementation, myometrial chemokine/cytokine and leukocyte numbers, but not Cx-43 and COX-2 expression, increased. These data show that during pregnancy P4 controls myometrial monocyte infiltration, cytokine and prolabor factor synthesis via mRNA-dependent and independent mechanisms and, with prolonged P4 supplementation, P4 action is repressed resulting in increased myometrial inflammation.


Asunto(s)
Miometrio/efectos de los fármacos , Parto/efectos de los fármacos , Progesterona/farmacología , Animales , Quimiocinas/metabolismo , Conexina 43/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Mifepristona/farmacología , Monocitos/metabolismo , Miometrio/inmunología , Miometrio/metabolismo , Neutrófilos/metabolismo , Parto/inmunología , Parto/metabolismo
8.
Crit Care Med ; 45(8): e831-e839, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28426531

RESUMEN

OBJECTIVE: Obesity has a complex impact on acute respiratory distress syndrome patients, being associated with increased likelihood of developing the syndrome but reduced likelihood of dying. We propose that such observations are potentially explained by a model in which obesity influences the iatrogenic injury that occurs subsequent to intensive care admission. This study therefore investigated whether fat feeding protected mice from ventilator-induced lung injury. DESIGN: In vivo study. SETTING: University research laboratory. SUBJECTS: Wild-type C57Bl/6 mice or tumor necrosis factor receptor 2 knockout mice, either fed a high-fat diet for 12-14 weeks, or age-matched lean controls. INTERVENTIONS: Anesthetized mice were ventilated with injurious high tidal volume ventilation for periods up to 180 minutes. MEASUREMENTS AND MAIN RESULTS: Fat-fed mice showed clear attenuation of ventilator-induced lung injury in terms of respiratory mechanics, blood gases, and pulmonary edema. Leukocyte recruitment and activation within the lungs were not significantly attenuated nor were a host of circulating or intra-alveolar inflammatory cytokines. However, intra-alveolar matrix metalloproteinase activity and levels of the matrix metalloproteinase cleavage product soluble receptor for advanced glycation end products were significantly attenuated in fat-fed mice. This was associated with reduced stretch-induced CD147 expression on lung epithelial cells. CONCLUSIONS: Consumption of a high-fat diet protects mice from ventilator-induced lung injury in a manner independent of neutrophil recruitment, which we postulate instead arises through blunted up-regulation of CD147 expression and subsequent activation of intra-alveolar matrix metalloproteinases. These findings may open avenues for therapeutic manipulation in acute respiratory distress syndrome and could have implications for understanding the pathogenesis of lung disease in obese patients.


Asunto(s)
Dieta Alta en Grasa , Obesidad/fisiopatología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Animales , Análisis de los Gases de la Sangre , Citocinas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/fisiología , Neutrófilos/metabolismo , Obesidad/epidemiología , Edema Pulmonar/fisiopatología , Edema Pulmonar/prevención & control , Mecánica Respiratoria , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/epidemiología
9.
Front Immunol ; 8: 128, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28243236

RESUMEN

BACKGROUND: Tumor necrosis factor-α (TNF) is strongly implicated in the development of acute respiratory distress syndrome (ARDS), but its potential as a therapeutic target has been hampered by its complex biology. TNF signals through two receptors, p55 and p75, which play differential roles in pulmonary edema formation during ARDS. We have recently shown that inhibition of p55 by a novel domain antibody (dAb™) attenuated ventilator-induced lung injury. In the current study, we explored the efficacy of this antibody in mouse models of acid-induced lung injury to investigate the longer consequences of treatment. METHODS: We employed two acid-induced injury models, an acute ventilated model and a resolving spontaneously breathing model. C57BL/6 mice were pretreated intratracheally or intranasally with p55-targeting dAb or non-targeting "dummy" dAb, 1 or 4 h before acid instillation. RESULTS: Acid instillation in the dummy dAb group caused hypoxemia, increased respiratory system elastance, pulmonary inflammation, and edema in both the ventilated and resolving models. Pretreatment with p55-targeting dAb significantly attenuated physiological markers of ARDS in both models. p55-targeting dAb also attenuated pulmonary inflammation in the ventilated model, with signs that altered cytokine production and leukocyte recruitment persisted beyond the very acute phase. CONCLUSION: These results demonstrate that the p55-targeting dAb attenuates lung injury and edema formation in models of ARDS induced by acid aspiration, with protection from a single dose lasting up to 24 h. Together with our previous data, the current study lends support toward the clinical targeting of p55 for patients with, or at risk of ARDS.

10.
Thorax ; 71(11): 1020-1029, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27287089

RESUMEN

BACKGROUND: Microvesicles (MVs) are important mediators of intercellular communication, packaging a variety of molecular cargo. They have been implicated in the pathophysiology of various inflammatory diseases; yet, their role in acute lung injury (ALI) remains unknown. OBJECTIVES: We aimed to identify the biological activity and functional role of intra-alveolar MVs in ALI. METHODS: Lipopolysaccharide (LPS) was instilled intratracheally into C57BL/6 mice, and MV populations in bronchoalveolar lavage fluid (BALF) were evaluated. BALF MVs were isolated 1 hour post LPS, assessed for cytokine content and incubated with murine lung epithelial (MLE-12) cells. In separate experiments, primary alveolar macrophage-derived MVs were incubated with MLE-12 cells or instilled intratracheally into mice. RESULTS: Alveolar macrophages and epithelial cells rapidly released MVs into the alveoli following LPS. At 1 hour, the dominant population was alveolar macrophage-derived, and these MVs carried substantive amounts of tumour necrosis factor (TNF) but minimal amounts of IL-1ß/IL-6. Incubation of these mixed MVs with MLE-12 cells induced epithelial intercellular adhesion molecule-1 (ICAM-1) expression and keratinocyte-derived cytokine release compared with MVs from untreated mice (p<0.001). MVs released in vitro from LPS-primed alveolar macrophages caused similar increases in MLE-12 ICAM-1 expression, which was mediated by TNF. When instilled intratracheally into mice, these MVs induced increases in BALF neutrophils, protein and epithelial cell ICAM-1 expression (p<0.05). CONCLUSIONS: We demonstrate, for the first time, the sequential production of MVs from different intra-alveolar precursor cells during the early phase of ALI. Our findings suggest that alveolar macrophage-derived MVs, which carry biologically active TNF, may play an important role in initiating ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/fisiopatología , Micropartículas Derivadas de Células/metabolismo , Macrófagos Alveolares/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Citocinas/metabolismo , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL
11.
Crit Care Med ; 43(7): 1375-85, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25867908

RESUMEN

OBJECTIVES: To determine the effect of severe sepsis on monocyte tumor necrosis factor-α-converting enzyme baseline and inducible activity profiles. DESIGN: Observational clinical study. SETTING: Mixed surgical/medical teaching hospital ICU. PATIENTS: Sixteen patients with severe sepsis, 15 healthy volunteers, and eight critically ill patients with noninfectious systemic inflammatory response syndrome. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Monocyte expression of human leukocyte antigen-D-related peptide, sol-tumor necrosis factor production, tumor necrosis factor-α-converting enzyme expression and catalytic activity, tumor necrosis factor receptor 1 and 2 expression, and shedding at 48-hour intervals from day 0 to day 4, as well as p38-mitogen activated protein kinase expression. Compared with healthy volunteers, both sepsis and systemic inflammatory response syndrome patients' monocytes expressed reduced levels of human leukocyte antigen-D-related peptide and released less sol-tumor necrosis factor on in vitro lipopolysaccharide stimulation, consistent with the term monocyte deactivation. However, patients with sepsis had substantially elevated levels of basal tumor necrosis factor-α-converting enzyme activity that were refractory to lipopolysaccharide stimulation and this was accompanied by similar changes in p38-mitogen activated protein kinase signaling. In patients with systemic inflammatory response syndrome, monocyte basal tumor necrosis factor-α-converting enzyme, and its induction by lipopolysaccharide, appeared similar to healthy controls. Changes in basal tumor necrosis factor-α-converting enzyme activity at day 0 for sepsis patients correlated with Acute Physiology and Chronic Health Evaluation II score and the attenuated tumor necrosis factor-α-converting enzyme response to lipopolysaccharide was associated with increased mortality. Similar changes in monocyte tumor necrosis factor-α-converting enzyme activity could be induced in healthy volunteer monocytes using an in vitro two-hit inflammation model. Patients with sepsis also displayed reduced shedding of monocyte tumor necrosis factor receptors upon stimulation with lipopolysaccharide. CONCLUSIONS: Monocyte tumor necrosis factor-α-converting enzyme catalytic activity appeared altered by sepsis and may result in reduced shedding of tumor necrosis factor receptors. Changes seemed specific to sepsis and correlated with illness severity. A better understanding of how tumor necrosis factor-α-converting enzyme function is altered during sepsis will enhance our understanding of sepsis pathophysiology, which will help in the assessment of patient inflammatory status and ultimately may provide new strategies to treat sepsis.


Asunto(s)
Proteínas ADAM/fisiología , Inflamación/sangre , Monocitos/enzimología , Sepsis/sangre , Proteína ADAM17 , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad
12.
Ann Surg ; 259(6): 1235-44, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24263322

RESUMEN

OBJECTIVE: Postoperative cognitive decline is emerging as a significant complication of surgery among older adults. Animal models indicate a central role of hippocampal inflammatory responses in the pathophysiology of postoperative cognitive decline. We hypothesized that atorvastatin, shown to exert neuroprotective potential in central nervous system (CNS) disorders, would attenuate neuroinflammation and improve cognitive function in mice after surgery and anesthesia. METHODS: C57BL6 adult mice were pretreated with atorvastatin (250 µg) or vehicle, orally, for 5 days before undergoing unilateral nephrectomy under isoflurane anesthesia. We evaluated behavioral parameters related to cognitive function (fear conditioning and Morris Water Maze) and determined systemic and hippocampal interleukin-1ß levels, postoperatively. Endothelial COX-2 expression, gross NF-κB and microglial (IBA1, CD68) activation, synaptic function (synapsin-1, PSD95, COX-2), heme oxygenase-1, and GSK3ß were also examined. RESULTS: Surgery induced a significant reduction in hippocampal-dependent fear response that was attenuated by treatment with atorvastatin, which also preserved spatial memory on day 7 after surgery. Atorvastatin evoked significant protection from hippocampal interleukin-1ß production, but not systemic interleukin-1ß production, accompanied by a marked reduction in hippocampal endothelial COX-2, NF-κB activation and decreased microglial reactivity. Surgery triggered an acute decline in synapsin-1, paralleled by an increase in postsynaptic COX-2 that was partially attenuated by atorvastatin. Furthermore, phosphorylation and inactivation of neuronal GSK3ß was significantly enhanced after atorvastatin treatment. CONCLUSIONS: These findings indicate that cognitive decline is very likely associated with synaptic pathology after systemic and central inflammation induced by peripheral surgery/isoflurane anesthesia and suggest that the anti-inflammatory and neuroprotective properties of atorvastatin provide a rationale for its use as a therapeutic strategy for postoperative cognitive decline.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Cognición/efectos de los fármacos , Ácidos Heptanoicos/administración & dosificación , Memoria/efectos de los fármacos , Nefrectomía/efectos adversos , Pirroles/administración & dosificación , Recuperación de la Función/efectos de los fármacos , Administración Oral , Animales , Atorvastatina , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Complicaciones Posoperatorias , Resultado del Tratamiento
13.
Crit Care Med ; 42(1): e49-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23963135

RESUMEN

OBJECTIVES: Ventilator-induced lung injury has substantive impact on mortality of patients with acute respiratory distress syndrome. Although low tidal volume ventilation has been shown to reduce mortality, clinical benefits of open-lung strategy are controversial. In this study, we investigated the impact of two distinct forms of ventilator-induced lung injury, i.e., volutrauma and atelectrauma, on the progression of lung injury and inflammation, in particular alveolar and systemic cytokine production. DESIGN: Ex vivo study. SETTING: University research laboratory. SUBJECTS: C57BL/6 mice. INTERVENTIONS: Isolated, buffer-perfused lungs were allocated to one of three ventilatory protocols for 3 hours: control group received low tidal volume (7 mL/kg) with positive end-expiratory pressure (5 cm H2O) and regular sustained inflation; high-stretch group received high tidal volume (30-32 mL/kg) with positive end-expiratory pressure (3 cm H2O) and sustained inflation; and atelectasis group received the same tidal volume as control but neither positive end-expiratory pressure nor sustained inflation. MEASUREMENTS AND MAIN RESULTS: Both injurious ventilatory protocols developed comparable levels of physiological injury and pulmonary edema, measured by respiratory system mechanics and lavage fluid protein. High-stretch induced marked increases in proinflammatory cytokines in perfusate and lung lavage fluid, compared to control. In contrast, atelectasis had no effect on perfusate cytokines compared to control but did induce some up-regulation of lavage cytokines. Depletion of monocytes marginated within the lung microvasculature, achieved by pretreating mice with i.v. liposome-encapsulated clodronate, significantly attenuated perfusate cytokine levels, especially tumor necrosis factor, in the high-stretch, but not atelectasis group. CONCLUSIONS: Volutrauma (high-stretch), but not atelectrauma (atelectasis), directly activates monocytes within the pulmonary vasculature, leading to cytokine release into systemic circulation. We postulate this as a potential explanation why open-lung strategy has limited mortality benefits in ventilated critically ill patients.


Asunto(s)
Citocinas/fisiología , Monocitos/fisiología , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Animales , Citocinas/biosíntesis , Edema/etiología , Edema/inmunología , Edema/fisiopatología , Citometría de Flujo , Pulmón/citología , Pulmón/inmunología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Respiración con Presión Positiva/efectos adversos , Respiración con Presión Positiva/métodos , Volumen de Ventilación Pulmonar/fisiología , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología
14.
J Immunol ; 190(8): 4274-82, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23487422

RESUMEN

The ability of the alveolar epithelium to prevent and resolve pulmonary edema is a crucial determinant of morbidity and mortality in acute lung injury (ALI). TNF has been implicated in ALI pathogenesis, but the precise mechanisms remain undetermined. We evaluated the role of TNF signaling in pulmonary edema formation in a clinically relevant mouse model of ALI induced by acid aspiration and investigated the effects of TNF p55 receptor deletion, caspase-8 inhibition, and alveolar macrophage depletion on alveolar epithelial function. We found that TNF plays a central role in the development of pulmonary edema in ALI through activation of p55-mediated death signaling, rather than through previously well-characterized p55-mediated proinflammatory signaling. Acid aspiration produced pulmonary edema with significant alveolar epithelial dysfunction, as determined by alveolar fluid clearance (AFC) and intra-alveolar levels of the receptor for advanced glycation end-products. The impairment of AFC was strongly correlated with lung caspase-8 activation, which was localized to type 1 alveolar epithelial cells by flow cytometric analysis. p55-deficient mice displayed markedly attenuated injury, with improved AFC and reduced caspase-8 activity but no differences in downstream cytokine/chemokine production and neutrophil recruitment. Caspase-8 inhibition significantly improved AFC and oxygenation, whereas depletion of alveolar macrophages attenuated epithelial dysfunction with reduced TNF production and caspase-8 activity. These results provide in vivo evidence for a novel role for TNF p55 receptor-mediated caspase-8 signaling, without substantial apoptotic cell death, in triggering alveolar epithelial dysfunction and determining the early pathophysiology of ALI. Blockade of TNF-induced death signaling may provide an effective early-phase strategy for ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/toxicidad , Lesión Pulmonar Aguda/metabolismo , Animales , Caspasa 8/toxicidad , Muerte Celular/inmunología , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/toxicidad , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Edema Pulmonar/inmunología , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L812-21, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21873449

RESUMEN

Margination and activation of monocytes within the pulmonary microcirculation contribute substantially to the development of acute lung injury in mice. The enhanced LPS-induced TNF expression exhibited by Gr-1(high) compared with Gr-1(low) monocytes within the lung microvasculature suggests differential roles for these subsets. We investigated the mechanisms responsible for such heterogeneity of lung-marginated monocyte proinflammatory response using a combined in vitro and in vivo approach. The monocyte subset inflammatory response was studied in vitro in mouse peripheral blood mononuclear cell-lung endothelial cell coculture and in vivo in a two-hit model of intravenous LPS-induced monocyte margination and lung inflammation in mice, by flow cytometry-based quantification of proinflammatory genes and intracellular phospho-kinases. With LPS stimulation in vitro, TNF expression was consistently higher in Gr-1(high) than Gr-1(low) monocytes, markedly enhanced by coculture with endothelial cells, and abrogated by p38 MAPK inhibitors. Expression of IL-6, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) was only detectable under coculture conditions, was substantially higher in Gr-1(high) monocytes, and was attenuated by p38 inhibition. Consistent with these differential responses, phosphorylation of p38 and its substrate MAPK-activated protein kinase 2 (MK2) was significantly higher in the Gr-1(high) subset. In vivo, p38 inhibitor treatment significantly attenuated LPS-induced TNF expression in "lung-marginated" Gr-1(high) monocytes. LPS-induced p38/MK2 phosphorylation was higher in lung-marginated Gr-1(high) than Gr-1(low) monocytes and neutrophils, mirroring TNF expression. These results indicate that the p38/MK2 pathway is a critical determinant of elevated Gr-1(high) subset responsiveness within the lung microvasculature, producing a coordinated proinflammatory response that places Gr-1(high) monocytes as key orchestrators of pulmonary microvascular inflammation and injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/inmunología , Microvasos/inmunología , Monocitos/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/fisiopatología , Animales , Linaje de la Célula , Técnicas de Cocultivo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Citometría de Flujo , Inflamación/inmunología , Inflamación/fisiopatología , Interleucina-6/análisis , Interleucina-6/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Lipopolisacáridos/efectos adversos , Lipopolisacáridos/farmacología , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/metabolismo , Monocitos/citología , Monocitos/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
16.
J Biol Chem ; 286(41): 35466-35476, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21865167

RESUMEN

Tumor necrosis factor α-converting enzyme (TACE) is responsible for the shedding of cell surface TNF. Studies suggest that reactive oxygen species (ROS) mediate up-regulation of TACE activity by direct oxidization or modification of the protein. However, these investigations have been largely based upon nonphysiological stimulation of promonocytic cell lines which may respond and process TACE differently from primary cells. Furthermore, investigators have relied upon TACE substrate shedding as a surrogate for activity quantification. We addressed these concerns, employing a direct, cell-based fluorometric assay to investigate the regulation of TACE catalytic activity on freshly isolated primary human monocytes during LPS stimulation. We hypothesized that ROS mediate up-regulation of TACE activity indirectly, by activation of intracellular signaling pathways. LPS up-regulated TACE activity rapidly (within 30 min) without changing cell surface TACE expression. Scavenging of ROS or inhibiting their production by flavoprotein oxidoreductases significantly attenuated LPS-induced TACE activity up-regulation. Exogenous ROS (H(2)O(2)) also up-regulated TACE activity with similar kinetics and magnitude as LPS. H(2)O(2)- and LPS-induced TACE activity up-regulation were effectively abolished by a variety of selective p38 MAPK inhibitors. Activation of p38 was redox-sensitive as H(2)O(2) caused p38 phosphorylation, and ROS scavenging significantly reduced LPS-induced phospho-p38 expression. Inhibition of the p38 substrate, MAPK-activated protein kinase 2, completely attenuated TACE activity up-regulation, whereas inhibition of ERK had little effect. Lastly, inhibition of cell surface oxidoreductases prevented TACE activity up-regulation distal to p38 activation. In conclusion, our data indicate that in primary human monocytes, ROS mediate LPS-induced up-regulation of TACE activity indirectly through activation of the p38 signaling pathway.


Asunto(s)
Proteínas ADAM/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Monocitos/enzimología , Oxidantes/farmacología , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína ADAM17 , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Peróxido de Hidrógeno/metabolismo , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Monocitos/citología , Oxidantes/metabolismo , Regulación hacia Arriba/fisiología
17.
J Appl Physiol (1985) ; 111(1): 177-84, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21512145

RESUMEN

Elevated soluble tumor necrosis factor-α receptor (sTNFR) levels in bronchoalveolar lavage fluid (BALF) are associated with poor patient outcome in acute lung injury (ALI). The mechanisms underlying these increases are unknown, but it is possible that pulmonary inflammation and increased alveolar epithelial permeability may individually contribute. We investigated mechanisms of elevated BALF sTNFRs in two in vivo mouse models of ALI. Anesthetized mice were challenged with intratracheal lipopolysaccharide or subjected to injurious mechanical ventilation. Lipopolysaccharide instillation produced acute intra-alveolar inflammation, but minimal alveolar epithelial permeability changes, with increased BALF sTNFR p75, but not p55. Increased p75 levels were markedly attenuated by alveolar macrophage depletion. In contrast, injurious ventilation induced substantial alveolar epithelial permeability, with increased BALF p75 and p55, which strongly correlated with total protein. BALF sTNFRs were not increased in isolated buffer-perfused lungs (devoid of circulating sTNFRs) subjected to injurious ventilation. These results suggest that lipopolysaccharide-induced intra-alveolar inflammation upregulates alveolar macrophage-mediated production of sTNFR p75, whereas enhanced alveolar epithelial permeability following mechanical ventilation leads to increased BALF p75 and p55 via plasma leakage. These data provide new insights into differential regulation of intra-alveolar sTNFR levels during ALI and may suggest sTNFRs as potential markers for evaluating the pathophysiology of ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Neumonía/inmunología , Alveolos Pulmonares/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Permeabilidad Capilar , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Lipopolisacáridos , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía/etiología , Alveolos Pulmonares/irrigación sanguínea , Respiración Artificial/efectos adversos , Factores de Tiempo , Regulación hacia Arriba , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología
18.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L781-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21378027

RESUMEN

TNF plays a crucial role in the pathogenesis of acute lung injury. However, the expression profile of its two receptors, p55 and p75, on pulmonary endothelium and their influence on TNF signaling during lung microvascular inflammation remain uncertain. Using flow cytometry, we characterized the expression profile of TNF receptors on the surface of freshly harvested pulmonary endothelial cells (PECs) from mice and found expression of both receptors with dominance of p55. To investigate the impact of stimulating individual TNF receptors, we treated wild-type and TNF receptor knockout mice with intravenous TNF and determined surface expression of adhesion molecules (E-selectin, VCAM-1, ICAM-1) on PECs by flow cytometry. TNF-induced upregulation of all adhesion molecules was substantially attenuated by absence of p55, whereas lack of p75 had a similar but smaller effect that varied between adhesion molecules. Selective blockade of individual TNF receptors by specific antibodies in wild-type primary PEC culture confirmed that the in vivo findings were due to direct effects of TNF receptor inhibition on endothelium and not other cells (e.g., circulating leukocytes). Finally, we found that PEC surface expression of p55 dramatically decreased in the early stages of endotoxemia following intravenous LPS, while no change in p75 expression was detected. These data demonstrate a crucial in vivo role of p55 and an auxiliary role of p75 in TNF-mediated adhesion molecule upregulation on PECs. It is possible that the importance of the individual receptors varies at different stages of pulmonary microvascular inflammation following changes in their relative expression.


Asunto(s)
Células Endoteliales/metabolismo , Pulmón/fisiología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Receptores Señuelo del Factor de Necrosis Tumoral/metabolismo , Animales , Selectina E/metabolismo , Endotoxemia/fisiopatología , Molécula 1 de Adhesión Intercelular/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/fisiopatología , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/biosíntesis
19.
J Immunol ; 182(2): 1155-66, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124759

RESUMEN

The specialized role of mouse Gr-1(high) monocytes in local inflammatory reactions has been well documented, but the trafficking and responsiveness of this subset during systemic inflammation and their contribution to sepsis-related organ injury has not been investigated. Using flow cytometry, we studied monocyte subset margination to the pulmonary microcirculation during subclinical endotoxemia in mice and investigated whether marginated monocytes contribute to lung injury in response to further septic stimuli. Subclinical low-dose i.v. LPS induced a rapid (within 2 h), large-scale mobilization of bone marrow Gr-1high monocytes and their prolonged margination to the lungs. With secondary LPS challenge, membrane TNF expression on these premarginated monocytes substantially increased, indicating their functional priming in vivo. Zymosan challenge produced small increases in pulmonary vascular permeability, which were markedly enhanced by the preadministration of low-dose LPS. The LPS-zymosan-induced permeability increases were effectively abrogated by pretreatment (30 min before zymosan challenge) with the platelet-activating factor antagonist WEB 2086 in combination with the phosphatidylcholine-phospholipase C inhibitor D609, suggesting the involvement of platelet-activating factor/ceramide-mediated pathways in this model. Depletion of monocytes (at 18 h after clodronate-liposome treatment) significantly attenuated the LPS-zymosan-induced permeability increase. However, restoration of normal LPS-induced Gr-1high monocyte margination to the lungs (at 48 h after clodronate-liposome treatment) resulted in the loss of this protective effect. These results demonstrate that mobilization and margination of Gr-1high monocytes during subclinical endotoxemia primes the lungs toward further septic stimuli and suggest a central role for this monocyte subset in the development of sepsis-related acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/patología , Células de la Médula Ósea/patología , Movimiento Celular/inmunología , Endotoxemia/patología , Mediadores de Inflamación/toxicidad , Monocitos/patología , Receptores de Quimiocina/biosíntesis , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Permeabilidad Capilar/inmunología , Diferenciación Celular/inmunología , Relación Dosis-Respuesta Inmunológica , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Endotoxemia/inducido químicamente , Endotoxemia/inmunología , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Receptores de Quimiocina/sangre , Receptores de Quimiocina/fisiología , Sepsis/sangre , Sepsis/inmunología , Sepsis/patología , Zimosan/toxicidad
20.
Am J Physiol Lung Cell Mol Physiol ; 293(1): L60-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17435079

RESUMEN

Ventilator-induced lung injury plays a crucial role in the outcome of patients with acute lung injury. Previous studies have shown a role for the cytokine tumor necrosis factor-alpha (TNF) in stretch-induced alveolar neutrophil recruitment, but the involvement of TNF in stretch-induced pulmonary edema is unclear. We investigated the effects of TNF through its individual p55 and p75 receptors on early pulmonary edema formation during high stretch ventilation, before neutrophil infiltration. Anesthetized wild-type or TNF receptor single/double knockout mice were ventilated with high tidal volume ( approximately 38 ml/kg) for 2 h or until they developed arterial hypotension. Pulmonary edema was assessed by physiological parameters including respiratory mechanics and blood gases, and by lavage fluid protein, lung wet:dry weight ratio, and lung permeability measurements using fluorescence-labeled albumin. High stretch ventilation in wild-type and TNF receptor double knockout animals induced similar pulmonary edema, and only 25-30% of mice completed the protocol. In contrast, the p55 receptor knockout mice were strongly protected from edema formation, with all animals completing the protocol. Myeloperoxidase assay indicated that this protective effect was not associated with decreased pulmonary neutrophil sequestration. The p75 receptor knockout mice, however, displayed increased susceptibility to edema formation, and no animals survived the full 2 h. These results demonstrate a novel role for TNF signaling (independent from its effects on neutrophil recruitment) specifically through the p55 receptor, in promoting high stretch-induced pulmonary edema, whereas p75 signaling may play an opposing role.


Asunto(s)
Edema Pulmonar/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Resistencia de las Vías Respiratorias , Animales , Análisis de los Gases de la Sangre , Barrera Alveolocapilar/fisiopatología , Líquido del Lavado Bronquioalveolar/química , Rendimiento Pulmonar , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mortalidad , Permeabilidad , Peroxidasa/metabolismo , Neumonía/fisiopatología , Presión , Edema Pulmonar/sangre , Edema Pulmonar/fisiopatología , Ventilación Pulmonar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA