Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gastroenterology ; 149(7): 1920-1931.e8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26302488

RESUMEN

BACKGROUND & AIMS: Neutrophils are involved in the development of acute pancreatitis (AP), but it is not clear how neutrophil-induced tissue damage is regulated. In addition to secreting antimicrobial compounds, activated neutrophils eliminate invading microorganisms by expelling nuclear DNA and histones to form extracellular web-like structures called neutrophil extracellular traps (NETs). However, NETs have been reported to contribute to organ dysfunction in patients with infectious diseases. We investigated whether NETs contribute to the development of AP in mice. METHODS: AP was induced in C57BL/6 mice by infusion of taurocholate into the pancreatic duct or by intraperitoneal administration of L-arginine. Pancreata were collected and extracellular DNA was detected by Sytox green staining, levels of CXC chemokines, histones, and cytokines also were measured. Cell-free DNA was quantified in plasma samples. Signal transducer and activator of transcription 3 phosphorylation and trypsin activation were analyzed in isolated acinar cells. NETs were depleted by administration of DNase I to mice. Plasma was obtained from healthy individuals (controls) and patients with severe AP. RESULTS: Infusion of taurocholate induced formation of NETs in pancreatic tissues of mice and increased levels of cell-free DNA in plasma. Neutrophil depletion prevented taurocholate-induced deposition of NETs in the pancreas. Administration of DNase I to mice reduced neutrophil infiltration and tissue damage in the inflamed pancreas and lung, and decreased levels of blood amylase, macrophage inflammatory protein-2, interleukin 6, and high-mobility groups protein 1. In mice given taurocholate, DNase I administration also reduced expression of integrin α M (macrophage-1 antigen) on circulating neutrophils. Similar results occurred in mice with L-arginine-induced AP. Addition of NETs and histones to acinar cells induced formation of trypsin and activation of signal transducer and activator of transcription 3; these processes were blocked by polysialic acid. Patients with severe AP had increased plasma levels of NET components compared with controls. CONCLUSIONS: NETs form in the pancreata of mice during the development of AP, and NET levels are increased in plasma from patients with AP, compared with controls. NETs regulate organ inflammation and injury in mice with AP, and might be targeted to reduce pancreatic tissue damage and inflammation in patients.


Asunto(s)
Trampas Extracelulares/metabolismo , Mediadores de Inflamación/sangre , Neutrófilos/enzimología , Páncreas/enzimología , Pancreatitis/enzimología , Tripsina/metabolismo , Enfermedad Aguda , Animales , Arginina , Estudios de Casos y Controles , ADN/sangre , Desoxirribonucleasa I/farmacología , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Pulmón/enzimología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Infiltración Neutrófila , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/patología , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/patología , Pancreatitis/sangre , Pancreatitis/inducido químicamente , Pancreatitis/inmunología , Pancreatitis/patología , Pancreatitis/prevención & control , Índice de Severidad de la Enfermedad , Ácido Taurocólico
2.
Inflamm Res ; 65(5): 405-13, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26873877

RESUMEN

OBJECTIVE: Systemic inflammatory response syndrome is associated with severe coagulopathy. The purpose of this study was to examine thrombin generation in systemic inflammation triggered by the endotoxin lipopolysaccharide (LPS) and the exotoxin streptococcal M1 protein. METHODS: Thrombin generation, lung histology and myeloperoxidase (MPO) activity were determined 6 and 24 h after induction of systemic inflammation. Male C57BL/6 mice received the Rac1 inhibitor NSC23766 prior to challenge with bacterial toxins. RESULTS: LPS and M1 protein challenge increased neutrophil infiltration and caused damage in the lung. Time to peak thrombin formation was increased and peak and total generation of thrombin were decreased in plasma from LPS- and M1 protein-treated mice. Coincubation of samples from mice exposed to bacterial toxins with platelet poor plasma from healthy mice completely reversed the inhibitory effect of LPS and M1 protein on thrombin generation, suggesting that bacterial toxins decreased levels of plasma factors explaining the reduction of thrombin generating capacity of plasma from septic animals. NSC23766 treatment not only decreased LPS- and M1 protein-induced neutrophil accumulation as well as levels of interleukin-6 and CXCL2 in the lung, but also abolished bacterial toxin-induced changes in thrombin generation. For example, NSC23766 increased peak formation by 57% and total thrombin generation by 48% in LPS-treated animals at 6 h. CONCLUSIONS: Taken together, our novel findings show that bacterial toxins increase thrombin generation via consumption of plasma factors and that Rac1 signaling plays an important role in thrombin generation in response to bacterial toxins. Thus, targeting Rac1 activity might be a useful way not only to ameliorate pulmonary inflammation, but also inhibit pathological changes in coagulation in bacterial infections.


Asunto(s)
Antígenos Bacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/farmacología , Proteínas Portadoras/farmacología , Lipopolisacáridos/farmacología , Neuropéptidos/metabolismo , Trombina/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Animales , Quimiocina CXCL2/metabolismo , Interleucina-6/metabolismo , Recuento de Leucocitos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Neuropéptidos/antagonistas & inhibidores , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/metabolismo , Recuento de Plaquetas , Protrombina/análisis , Pirimidinas/farmacología , Proteína de Unión al GTP rac1/antagonistas & inhibidores
3.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1159-67, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840996

RESUMEN

Streptococcus pyogenes cause infections ranging from mild pharyngitis to severe streptococcal toxic shock syndrome (STSS). The M1 serotype of Streptococcus pyogenes is most frequently associated with STSS. Herein, it was hypothesized that STAT3 signaling might be involved in M1 protein-evoked lung inflammation. The STAT3 inhibitor, S3I-201, was administered to male C57Bl/6 mice before iv challenge with M1 protein. Bronchoalveolar fluid and lung tissue were harvested for quantification of STAT3 activity, neutrophil recruitment, edema, and CXC chemokine formation. Neutrophil expression of Mac-1 was quantified by use of flow cytometry. Levels of IL-6 and HMGB1 were determined in plasma. CXCL2-induced neutrophil chemotaxis was studied in vitro. Administration of S3I-201 markedly reduced M1 protein-provoked STAT3 activity, neutrophil recruitment, edema formation, and inflammatory changes in the lung. In addition, M1 protein significantly increased Mac-1 expression on neutrophils and CXC chemokine levels in the lung. Treatment with S3I-201 had no effect on M1 protein-induced expression of Mac-1 on neutrophils. In contrast, inhibition of STAT3 activity greatly reduced M1 protein-induced formation of CXC chemokines in the lung. Interestingly, STAT3 inhibition markedly decreased plasma levels of IL-6 and HMGB1 in animals exposed to M1 protein. Moreover, we found that S3I-201 abolished CXCL2-induced neutrophil migration in vitro. In conclusion, these novel findings indicate that STAT3 signaling plays a key role in mediating CXC chemokine production and neutrophil infiltration in M1 protein-induced acute lung inflammation.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Neutrófilos/fisiología , Factor de Transcripción STAT3/fisiología , Animales , Movimiento Celular , Proteína HMGB1/sangre , Interleucina-6/sangre , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones Endogámicos C57BL , Infiltración Neutrófila
4.
Lab Invest ; 94(9): 1054-63, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25046439

RESUMEN

Matrix metalloproteinase-9 (MMP-9) regulates platelet shedding of CD40L in abdominal sepsis. However, the signaling mechanisms controlling sepsis-induced shedding of CD40L from activated platelets remain elusive. Rac1 has been reported to regulate diverse functions in platelets; we hypothesized herein that Rac1 might regulate platelet shedding of CD40L in sepsis. The specific Rac1 inhibitor NSC23766 (N6-[2-[[4-(diethylamino)-1-methylbutyl] amino]-6-methyl-4-pyrimidinyl]-2 methyl-4, 6-quinolinediamine trihydrochloride) was administered to mice undergoing cecal ligation and puncture (CLP). Levels of CD40L and MMP-9 in plasma, platelets, and neutrophils were determined by use of ELISA, western blot, and confocal microscopy. Platelet depletion abolished the CLP-induced increase in plasma levels of CD40L. Rac1 activity was significantly increased in platelets from septic animals. Administration of NSC23766 abolished the CLP-induced enhancement of soluble CD40L levels in the plasma. Moreover, Rac1 inhibition completely inhibited proteinase-activated receptor-4-induced surface mobilization and secretion of CD40L in isolated platelets. CLP significantly increased plasma levels of MMP-9 and Rac1 activity in neutrophils. Treatment with NSC23766 markedly attenuated MMP-9 levels in the plasma from septic mice. In addition, Rac1 inhibition abolished chemokine-induced secretion of MMP-9 from isolated neutrophils. Finally, platelet shedding of CD40L was significantly reduced in response to stimulation with supernatants from activated MMP-9-deficient neutrophils compared with supernatants from wild-type neutrophils, indicating a direct role of neutrophil-derived MMP-9 in regulating platelet shedding of CD40L. Our novel data suggest that sepsis-induced platelet shedding of CD40L is dependent on Rac1 signaling. Rac1 controls surface mobilization of CD40L on activated platelets and MMP-9 secretion from neutrophils. Thus, our findings indicate that targeting Rac1 signaling might be a useful way to control pathologic elevations of CD40L in the systemic circulation in abdominal sepsis.


Asunto(s)
Abdomen , Plaquetas/metabolismo , Ligando de CD40/sangre , Sepsis/sangre , Proteína de Unión al GTP rac1/fisiología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Masculino , Metaloproteinasa 8 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/enzimología
5.
Am J Physiol Gastrointest Liver Physiol ; 307(9): G914-21, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25214403

RESUMEN

Severe acute pancreatitis (AP) is characterized by leukocyte infiltration and tissue injury. Herein, we wanted to examine the potential effects of thrombin-derived host defense peptides (TDPs) in severe AP. Pancreatitis was provoked by infusion of taurocholate into the pancreatic duct or by intraperitoneal administration of l-arginine in C57BL/6 mice. Animals were treated with the TDPs GKY20 and GKY25 or a control peptide WFF25 30 min before induction of AP. TDPs reduced blood amylase levels, neutrophil infiltration, hemorrhage, necrosis, and edema formation in the inflamed pancreas. Treatment with TDPs markedly attenuated the taurocholate-induced increase in plasma levels of CXCL2 and interleukin-6. Moreover, administration of TDPs decreased histone 3, histone 4, and myeloperoxidase levels in the pancreas in response to taurocholate challenge. Interestingly, administration of TDPs abolished neutrophil expression of Mac-1 in mice with pancreatitis. In addition, TDPs inhibited CXCL2-induced chemotaxis of isolated neutrophils in vitro. Fluorescent-labeled TDP was found to directly bind to isolated neutrophils. Finally, a beneficial effect of TDPs was confirmed in l-arginine-induced pancreatitis. Our novel results demonstrate that TDPs exert protective effects against pathological inflammation and tissue damage in AP. These findings suggest that TDPs might be useful in the management of patients with severe AP.


Asunto(s)
Infiltración Neutrófila , Pancreatitis Aguda Necrotizante/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Trombina/uso terapéutico , Secuencia de Aminoácidos , Animales , Quimiocina CXCL2/sangre , Histonas/genética , Histonas/metabolismo , Humanos , Interleucina-6/sangre , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Páncreas/inmunología , Páncreas/metabolismo , Páncreas/patología , Pancreatitis Aguda Necrotizante/inmunología , Pancreatitis Aguda Necrotizante/metabolismo , Fragmentos de Péptidos/química , Peroxidasa/genética , Peroxidasa/metabolismo , Trombina/química
6.
J Surg Res ; 183(2): 798-807, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23545410

RESUMEN

Excessive neutrophil recruitment is a major feature in septic lung damage although the signaling mechanisms behind pulmonary infiltration of neutrophils in sepsis remain elusive. In the present study, we hypothesized that Rac1 might play an important role in pulmonary neutrophil accumulation and tissue injury in abdominal sepsis. Male C57BL/6 mice were treated with Rac1 inhibitor NSC23766 (5 mg/kg) before cecal ligation and puncture (CLP). Bronchoalveolar lavage fluid and lung tissue were collected for the quantification of neutrophil recruitment and edema and CXC chemokine formation. Blood was collected for the determination of Mac-1 on neutrophils and proinflammatory compounds in plasma. Gene expression of CXC chemokines and tumor necrosis factor alpha was determined by quantitative reverse transcription-polymerase chain reaction in alveolar macrophages. Rac1 activity was increased in lungs from septic animals, and NSC23766 significantly decreased pulmonary activity of Rac1 induced by CLP. Administration of NSC23766 markedly reduced CLP-triggered neutrophil infiltration, edema formation, and tissue damage in the lung. Inhibition of Rac1 decreased CLP-induced neutrophil expression of Mac-1 and pulmonary formation of CXC chemokines. Moreover, NSC23766 abolished the sepsis-evoked elevation of messenger RNA levels of CXC chemokines and tumor necrosis factor alpha in alveolar macrophages. Rac1 inhibition decreased the CLP-induced increase in plasma levels of high mobility group protein B1 and interleukin 6, indicating a role of Rac1 in systemic inflammation. In conclusion, our results demonstrate that Rac1 signaling plays a key role in regulating pulmonary infiltration of neutrophils and tissue injury via regulation of chemokine production in the lung and Mac-1 expression on neutrophils in abdominal sepsis. Thus, targeting Rac1 activity might be a useful strategy to protect the lung in abdominal sepsis.


Asunto(s)
Quimiocinas CXC/metabolismo , Antígeno de Macrófago-1/metabolismo , Neuropéptidos/fisiología , Neumonía/microbiología , Neumonía/fisiopatología , Sepsis/complicaciones , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/fisiología , Aminoquinolinas/farmacología , Animales , Ciego/lesiones , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Proteína HMGB1/metabolismo , Técnicas In Vitro , Ligadura/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Neumonía/patología , Punciones/efectos adversos , Pirimidinas/farmacología , Sepsis/etiología , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/efectos de los fármacos , Proteína de Unión al GTP rac1
7.
Int J Immunopathol Pharmacol ; 36: 3946320221085465, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35341332

RESUMEN

BACKGROUND: COVID-19 primarily presents as a respiratory tract infection, but studies indicate that it could be considered a systemic disease that can spread to affect multiple organ systems, including respiratory, cardiovascular, gastrointestinal, hematopoietic, neurological, and immune systems. OBJECTIVE: To describe and analyze the clinical and hematological characteristics of 300 hospitalized COVID-19 patients in Erbil, Kurdistan. METHODS: This retrospective study included 300 patients of any age admitted to hospital due to confirmed COVID-19 between September 2020 and February 2021. Cases were diagnosed by reverse transcriptase polymerase chain reaction assays of nasopharyngeal swab specimens. RESULTS: The highest proportion of patients were aged 21-40 years. The most common symptoms among the patients were myalgia (66.7%), fatigue (62.3%), headache (50.7%), and chest pain (52.7%). Differences in hematological and biochemical parameters were observed between deceased and recovered patients. Only the mid-range absolute count percentage (MID%) was significantly higher in the recovered patients than in the deceased ones (6.41% vs. 4.48, p = 0.019). Death was significantly higher among older patients (>40 years) than younger ones (≤40 years) (6.8% vs. 1.3%, p = 0.015), diabetic than non-diabetic (10.8% vs. 3%, p = 0.047), and those having chronic diseases than those without chronic diseases (10.6% vs. 2.1%, p = 0.006). CONCLUSIONS: Different hematological and biochemical parameter findings were observed among the COVID-19 patients. Low MID%, older age, and presence of diabetes mellitus and chronic disease were significantly associated with death among COVID-19 patients.


Asunto(s)
COVID-19 , Adulto , Hospitalización , Humanos , Irak/epidemiología , Estudios Retrospectivos , Adulto Joven
8.
Int Immunopharmacol ; 97: 107701, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33930704

RESUMEN

SARS-CoV-2 or Coronavirus disease 2019 (COVID-19) outbreak which caused by the severe acute respiratory syndrome, has rapidly spread over the world. The exact mechanism how this virus will affect the liver remained elusive. The aim of this study was to evaluate the liver function in patients with severe acute respiratory syndrome coronavirus 2 and potential causes of hepatic enzymes disease in these patients. Clinical characteristics and laboratory findings were collected from patients with COVID-19 who were admitted to the corona center in Erbil city/Kurdistan region of Iraq, from March 10 to July 10, 2020. Serum was collected from patients with COVID-19 and liver enzyme tests were measured. Liver alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and total bilirubin (TBIL) were analyzed in these patients. Of the 74 patients, 25 (34.7%) had abnormal ALT activity, 28 (40%) had abnormal AST activity, 12 (20.3%) had abnormal ALP activity, and 39 (52.7%) had abnormal total bilirubin P-value < 0.05. The inflammatory biomarkers CRP and IL-6 in COVID-19 patients with abnormal liver function test (4.9 ± 1.0 mg/dl) and (231.2 ± 35.7 pg/ml) respectively. The levels of both biomarkers were statistically significantly higher than COVID-19 patients with normal liver function test (2.1 ± 0.5 mg/dl) and (2.1 ± 0.5 mg/dl) respectively, P-value < 0.05. However, CRP and IL-6 were not statistically significant different between male and female COVID-19 patients P-value < 0.05. In conclusion, we found that most of the patients with SARS-CoV-2 have abnormal hepatic enzyme activities and that is might related to virus replication in the liver.


Asunto(s)
COVID-19/enzimología , COVID-19/virología , Hígado/enzimología , Hígado/virología , Adolescente , Adulto , Anciano , Alanina Transaminasa/sangre , Fosfatasa Alcalina/sangre , Aspartato Aminotransferasas/sangre , Bilirrubina/sangre , Biomarcadores/sangre , COVID-19/sangre , COVID-19/complicaciones , Portador Sano/sangre , Niño , Femenino , Humanos , Interleucina-6/sangre , Hepatopatías/sangre , Hepatopatías/enzimología , Hepatopatías/etiología , Hepatopatías/virología , Pruebas de Función Hepática , Masculino , Persona de Mediana Edad , Receptores Inmunológicos/sangre , Adulto Joven
9.
PLoS One ; 16(4): e0250330, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33914789

RESUMEN

The emergence of the novel coronavirus and then pandemic outbreak was coined 2019- nCoV or COVID-19 (or SARS-CoV-2 disease 2019). This disease has a mortality rate of about 3·7 percent, and successful therapy is desperately needed to combat it. The exact cellular mechanisms of COVID-19 need to be illustrated in detail. This study aimed to evaluate serum cytokines in COVID-19 patients. In this study, serum was collected from volunteer individuals, moderate COVID-19 patients, severe cases of COVID-19 patients, and patients who recovered from COVID-19 (n = 122). The serum concentrations of interleukins such as IL-1, IL-4, IL-6, IL-8, IL-10, and tumor necrosis factor-alpha (TNF-α), were measured by enzyme-linked immunosorbent assays (ELISA). The concentrations of IL-1 and TNF-α were did not differ significantly among groups. However, the concentration of IL-6 was significantly higher in moderate COVID-19 and severe cases of COVID-19 groups compared to control and recovered groups indicating it to be an independent predictor in the coronavirus disease. The levels of IFN-γ and IL-4 were significantly lower in the recovery group than the severe case of the COVID-19 group. In contrast, the level of IL-10 in recovered COVID-19 patients was significantly higher in compare to severe cases, COVID-19 patients. Varying levels of cytokines were detected in COVID-19 group than control group suggesting distinct immunoregulatory mechanisms involved in COVID-19 pathogenesis. However, additional investigations are needed to be to be performed to understand the exact cellular mechanism of this disease.


Asunto(s)
COVID-19/sangre , Citocinas/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/epidemiología , Femenino , Humanos , Interferón gamma/sangre , Interleucina-10/sangre , Interleucina-4/sangre , Interleucina-6/sangre , Irak/epidemiología , Masculino , Persona de Mediana Edad , SARS-CoV-2/aislamiento & purificación , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
10.
Br J Pharmacol ; 172(22): 5347-59, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26478565

RESUMEN

BACKGROUND AND PURPOSE: Platelets are potent regulators of neutrophil accumulation in septic lung damage. We hypothesized that platelet-derived CXCL4 might support pulmonary neutrophilia in a murine model of abdominal sepsis. EXPERIMENTAL APPROACH: Polymicrobial sepsis was triggered by coecal ligation and puncture (CLP) in C57BL/6 mice. Platelet secretion of CXCL4 was studied by using confocal microscopy. Plasma and lung levels of CXCL4, CXCL1 and CXCL2 were determined by elisa. Flow cytometry was used to examine surface expression of Mac-1 on neutrophils. KEY RESULTS: CLP increased CXCL4 levels in plasma, and platelet depletion reduced plasma levels of CXCL4 in septic animals. Rac1 inhibitor NSC23766 decreased the CLP-enhanced CXCL4 in plasma by 77%. NSC23766 also abolished PAR4 agonist-induced secretion of CXCL4 from isolated platelets. Inhibition of CXCL4 reduced CLP-evoked neutrophil recruitment, oedema formation and tissue damage in the lung. However, immunoneutralization of CXCL4 had no effect on CLP-induced expression of Mac-1 on neutrophils. Targeting CXCL4 attenuated plasma and lung levels of CXCL1 and CXCL2 in septic mice. CXCL4 had no effect on neutrophil chemotaxis in vitro, indicating it has an indirect effect on pulmonary neutrophilia. Intratracheal CXCL4 enhanced infiltration of neutrophils and formation of CXCL2 in the lung. CXCR2 antagonist SB225002 markedly reduced CXCL4-provoked neutrophil accumulation in the lung. CXCL4 caused secretion of CXCL2 from isolated alveolar macrophages. CONCLUSIONS AND IMPLICATIONS: Rac1 controls platelet secretion of CXCL4 and CXCL4 is a potent stimulator of neutrophil accumulation in septic lungs via generation of CXCL2 in alveolar macrophages. Platelet-derived CXCL4 plays an important role in lung inflammation and tissue damage in polymicrobial sepsis.


Asunto(s)
Plaquetas/metabolismo , Pulmón/metabolismo , Neuropéptidos/metabolismo , Factor Plaquetario 4/metabolismo , Sepsis/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CXCL1/sangre , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/sangre , Quimiocina CXCL2/metabolismo , Pulmón/patología , Macrófagos Alveolares/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuropéptidos/sangre , Infiltración Neutrófila , Factor Plaquetario 4/sangre , Sepsis/patología , Proteína de Unión al GTP rac1/sangre
11.
J Leukoc Biol ; 97(5): 975-984, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25717148

RESUMEN

Accumulating evidence suggest that platelets play an important role in regulating neutrophil recruitment in septic lung injury. Herein, we hypothesized that platelet-derived CCL5 might facilitate sepsis-induced neutrophil accumulation in the lung. Abdominal sepsis was induced by CLP in C57BL/6 mice. CLP increased plasma levels of CCL5. Platelet depletion and treatment with the Rac1 inhibitor NSC23766 markedly reduced CCL5 in the plasma of septic mice. Moreover, Rac1 inhibition completely inhibited proteasePAR4-induced secretion of CCL5 in isolated platelets. Immunoneutralization of CCL5 decreased CLP-induced neutrophil infiltration, edema formation, and tissue injury in the lung. However, inhibition of CCL5 function had no effect on CLP-induced expression of Mac-1 on neutrophils. The blocking of CCL5 decreased plasma and lung levels of CXCL1 and CXCL2 in septic animals. CCL5 had no effect on neutrophil chemotaxis in vitro, suggesting an indirect effect of CCL5 on neutrophil recruitment. Intratracheal challenge with CCL5 increased accumulation of neutrophils and formation of CXCL2 in the lung. Administration of the CXCR2 antagonist SB225002 abolished CCL5-induced pulmonary recruitment of neutrophils. Isolated alveolar macrophages expressed significant levels of the CCL5 receptors CCR1 and CCR5. In addition, CCL5 triggered significant secretion of CXCL2 from isolated alveolar macrophages. Notably, intratracheal administration of clodronate not only depleted mice of alveolar macrophages but also abolished CCL5-induced formation of CXCL2 in the lung. Taken together, our findings suggest that Rac1 regulates platelet secretion of CCL5 and that CCL5 is a potent inducer of neutrophil recruitment in septic lung injury via formation of CXCL2 in alveolar macrophages.

12.
Eur J Pharmacol ; 733: 45-53, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24704370

RESUMEN

Streptococcal toxic shock syndrome (STSS) is associated with a high mortality rate. The M1 serotype of Streptococcus pyogenes is most frequently associated with STSS. Herein, we examined the role of Ras signaling in M1 protein-induced lung injury. Male C57BL/6 mice received the Ras inhibitor (farnesylthiosalicylic acid, FTS) prior to M1 protein challenge. Bronchoalveolar fluid and lung tissue were harvested for quantification of neutrophil recruitment, edema and CXC chemokine formation. Neutrophil expression of Mac-1 was quantified by use of flow cytometry. Quantitative RT-PCR was used to determine gene expression of CXC chemokines in alveolar macrophages. Administration of FTS reduced M1 protein-induced neutrophil recruitment, edema formation and tissue damage in the lung. M1 protein challenge increased Mac-1 expression on neutrophils and CXC chemokine levels in the lung. Inhibition of Ras activity decreased M1 protein-induced expression of Mac-1 on neutrophils and secretion of CXC chemokines in the lung. Moreover, FTS abolished M1 protein-provoked gene expression of CXC chemokines in alveolar macrophages. Ras inhibition decreased chemokine-mediated neutrophil migration in vitro. Taken together, our novel findings indicate that Ras signaling is a potent regulator of CXC chemokine formation and neutrophil infiltration in the lung. Thus, inhibition of Ras activity might be a useful way to antagonize streptococcal M1 protein-triggered acute lung injury.


Asunto(s)
Antígenos Bacterianos/toxicidad , Proteínas de la Membrana Bacteriana Externa/toxicidad , Proteínas Portadoras/toxicidad , Quimiocinas CXC/inmunología , Lesión Pulmonar/inducido químicamente , Macrófagos Alveolares/inmunología , Proteínas ras/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocinas CXC/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Farnesol/análogos & derivados , Farnesol/farmacología , Citometría de Flujo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Macrófagos Alveolares/metabolismo , Masculino , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Edema Pulmonar/inducido químicamente , Edema Pulmonar/inmunología , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Salicilatos/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas ras/antagonistas & inhibidores
13.
Pancreas ; 43(3): 427-35, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24622074

RESUMEN

OBJECTIVES: The signaling mechanisms controlling organ damage in the pancreas in severe acute pancreatitis (AP) remain elusive. Herein, we examined the role of farnesyltransferase signaling in AP. METHODS: Pancreatitis was provoked by the infusion of taurocholate into the pancreatic duct in C57BL/6 mice. Animals were treated with a farnesyltransferase inhibitor FTI-277 (25 mg/kg) before pancreatitis induction. RESULTS: FTI-277 decreased the blood amylase levels, pancreatic neutrophil infiltration, hemorrhage, and edema formation in the pancreas in mice challenged with taurocholate. Farnesyltransferase inhibition reduced the myeloperoxidase levels in the pancreas and lungs in response to taurocholate infusion. However, FTI-277 had no effect on the taurocholate-provoked formation of macrophage inflammatory protein-2 in the pancreas. Interestingly, farnesyltransferase inhibition abolished the neutrophil expression of macrophage-1 antigen in mice with pancreatitis. In addition, FTI-277 decreased the taurocholate-induced activation of the rat sarcoma protein in the pancreas. An important role of farnesyltransferase was confirmed in L-arginine-induced pancreatitis. CONCLUSIONS: These results demonstrate that farnesyltransferase signaling plays a significant role in AP by regulating neutrophil infiltration and tissue injury via the neutrophil expression of macrophage-1 antigen. Thus, our findings not only elucidate novel signaling mechanisms in pancreatitis but also suggest that farnesyltransferase might constitute a target in the management of severe AP.


Asunto(s)
Farnesiltransferasa/metabolismo , Infiltración Neutrófila/fisiología , Pancreatitis/enzimología , Transducción de Señal/fisiología , Células Acinares/efectos de los fármacos , Células Acinares/enzimología , Enfermedad Aguda , Amilasas/sangre , Animales , Arginina , Células Cultivadas , Quimiocina CXCL2/metabolismo , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Antígeno de Macrófago-1/metabolismo , Masculino , Metionina/análogos & derivados , Metionina/farmacología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Páncreas/efectos de los fármacos , Páncreas/enzimología , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/inmunología , Peroxidasa/metabolismo , Transducción de Señal/efectos de los fármacos , Ácido Taurocólico , Tripsina/metabolismo , Tripsinógeno/metabolismo
14.
Eur J Pharmacol ; 741: 90-6, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25084221

RESUMEN

Excessive neutrophil recruitment in the colon is a major feature in acute colitis although the signaling mechanisms behind colonic recruitment of neutrophils remain elusive. Herein, we hypothesized that Rac1 activity might play an important role in neutrophil infiltration in the inflamed colon. Female Balb/c mice were treated with the Rac1 inhibitor NSC23766 (0.5 and 5mg/kg) before and daily after administration of 5% dextran sodium sulfate (DSS). Colonic tissue was collected for quantification of neutrophil recruitment, interleukin-6 (IL-6) and CXC chemokine formation as well as histological damage score five days after challenge with DSS. Rac1 activity was determined by western blot and Mac-1 expression by flow cytometry in neutrophils. Administration of NSC23766 decreased DSS-induced neutrophil recruitment and tissue damage in the colon. Rac1 inhibition decreased colonic formation of IL-6 and CXC chemokines in experimental colitis. Chemokine challenge increased Rac1 activity in neutrophils and NSC23766 markedly reduced this neutrophil activity of Rac1. Inhibition of Rac1 abolished CXC chemokine-induced neutrophil chemotaxis and up-regulation of Mac-1 in vitro. Taken together, Rac1 signaling plays a significant role in controlling accumulation of neutrophils and tissue injury in experimental colitis. Thus, our novel results suggest that targeting Rac1 signaling might be a useful way to protect against neutrophil-mediated tissue injury in acute colitis.


Asunto(s)
Colitis/metabolismo , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/fisiología , Infiltración Neutrófila/fisiología , Neutrófilos/metabolismo , Transducción de Señal/fisiología , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/fisiología , Aminoquinolinas/farmacología , Animales , Colitis/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA