Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Ann Surg ; 260(6): 1112-20, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24646554

RESUMO

OBJECTIVE: To test whether the mucus layer, luminal digestive enzymes, and intestinal mast cells are critical components in the pathogenesis of trauma shock-induced gut and lung injury. BACKGROUND: Gut origin sepsis studies have highlighted the importance of the systemic component (ischemia-reperfusion) of gut injury, whereas the intraluminal component is less well studied. METHODS: In rats subjected to trauma hemorrhagic shock (T/HS) or sham shock, the role of pancreatic enzymes in gut injury was tested by diversion of pancreatic enzymes via pancreatic duct exteriorization whereas the role of the mucus layer was tested via the enteral administration of a mucus surrogate. In addition, the role of mast cells was assessed by measuring mast cell activation and the ability of pharmacologic inhibition of mast cells to abrogate gut and lung injury. Gut and mucus injury was characterized functionally, morphologically, and chemically. RESULTS: Pancreatic duct exteriorization abrogated T/HS-induced gut barrier loss and limited chemical mucus changes. The mucus surrogate prevented T/HS-induced gut and lung injury. Finally, pancreatic enzyme-induced gut and lung injury seems to involve mast cell activation because T/HS activates mast cells and pharmacologic inhibition of intestinal mast cells prevented T/HS-induced gut and lung injury. CONCLUSIONS: These results indicate that gut and gut-induced lung injury after T/HS involves a complex process consisting of intraluminal digestive enzymes, the unstirred mucus layer, and a systemic ischemic-reperfusion injury. This suggests the possibility of intraluminal therapeutic strategies.


Assuntos
Lesão Pulmonar Aguda/terapia , Enzimas/metabolismo , Intestinos/enzimologia , Choque Hemorrágico/terapia , Ferimentos e Lesões/complicações , Lesão Pulmonar Aguda/etiologia , Animais , Modelos Animais de Doenças , Mucosa Intestinal/enzimologia , Masculino , Elastase Pancreática/metabolismo , Ratos , Ratos Sprague-Dawley , Choque Hemorrágico/etiologia
2.
Crit Care Med ; 42(3): e200-10, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24317495

RESUMO

OBJECTIVE: Microvascular dysfunction is a key element in the development of the multiple organ dysfunction syndrome. Although the mechanisms for this response are unclear, RBC adhesion to endothelium may initiate intravascular occlusion leading to ischemic tissue injury. Thus, we tested the hypothesis that trauma-hemorrhage induces RBC-endothelial cell adhesion. DESIGN: Prospective in vivo and in vitro animal study and analysis of patient blood samples. SETTING: University research laboratory and hospital emergency and trauma units. INTERVENTION: We initially assayed RBC adhesion to endothelial cells in vitro using RBCs obtained from rats subjected to trauma-hemorrhagic shock or sham shock as well as from severely injured trauma patients. Subsequently, we measured the role of putative RBCs and endothelial cell receptors in the increased RBC-endothelial cell adhesive response. MAIN RESULTS: In both rats and humans, trauma-hemorrhagic shock increased RBC adhesion to endothelium as well as increasing several putative RBC surface adhesion molecules including CD36. The critical factor leading to RBC-endothelial cell adhesion was increased surface RBC CD36 expression. Adhesion of trauma-hemorrhagic shock RBCs was mediated, at least in part, by the binding of RBC CD36 to its cognate endothelial receptors (αVß3 and VCAM-1). Gut-derived factors carried in the intestinal lymphatics triggered these trauma-hemorrhagic shock-induced RBC changes because 1) preventing trauma-hemorrhagic shock intestinal lymph from reaching the systemic circulation abrogated the RBC effects, 2) in vitro incubation of naïve whole blood with trauma-hemorrhagic shock lymph replicated the in vivo trauma-hemorrhagic shock-induced RBC changes while 3) injection of trauma-hemorrhagic shock lymph into naïve animals recreated the RBC changes observed after actual trauma-hemorrhagic shock. CONCLUSIONS: 1) Trauma-hemorrhagic shock induces rapid RBC adhesion to endothelial cells in patients and animals. 2) Increased RBC CD36 expression characterizes the RBC-adhesive phenotype. 3) The RBC phenotypic and functional changes were induced by gut-derived humoral factors. These novel findings may explain the microvascular dysfunction occurring after trauma-hemorrhagic shock, sepsis, and other stress states.


Assuntos
Antígenos CD36/genética , Eritrócitos/citologia , Insuficiência de Múltiplos Órgãos/genética , Choque Traumático/genética , Animais , Antígenos CD36/metabolismo , Adesão Celular/genética , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Eritrócitos/fisiologia , Regulação da Expressão Gênica , Humanos , Técnicas In Vitro , Masculino , Insuficiência de Múltiplos Órgãos/fisiopatologia , Fenótipo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Estudos de Amostragem , Sensibilidade e Especificidade , Choque Hemorrágico/genética , Choque Hemorrágico/metabolismo , Choque Hemorrágico/fisiopatologia , Choque Traumático/metabolismo , Choque Traumático/fisiopatologia
3.
J Surg Res ; 187(1): 270-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24321622

RESUMO

BACKGROUND: Lactoferrin (LF) is a pleiotropic glycoprotein that is found in bodily secretions and is postulated to enhance the gastrointestinal barrier and promote mucosal immunity. Thus, the ability of talactoferrin, an oral recombinant form of human LF, to limit gut injury and the production of biologically active gut-derived products was tested using a rat model of trauma-hemorrhagic shock (T/HS). METHODS: Male rats were orally dosed with vehicle or talactoferrin (1000 mg/kg, every day) for 5 d before being subjected to T/HS or trauma-sham shock (T/SS). Subsequently, rats were subjected to a laparotomy (trauma) and hemorrhagic shock (mean arterial pressure, 30-35 mm Hg × 90 min) or to T/SS, followed by resuscitation with their shed blood. Before inducing shock, the mesenteric lymphatic duct was catheterized for collection of mesenteric lymph. Four hours after the end of the shock or sham-shock period, rats were sacrificed, a segment of the distal ileum was collected for morphologic analysis, and lymph samples were processed and frozen. Subsequently, lymph samples were tested in several pharmacodynamic assays, including endothelial cell permeability, neutrophil respiratory burst activity, and red blood cell (RBC) deformability. Total white blood cell counts in lymph samples were also quantified. RESULTS: Pretreatment with talactoferrin reduced the incidence of T/HS-induced morphologic injury of ileum to T/SS levels. Post-T/HS lymph from vehicle-treated rats increased endothelial monolayer permeability and neutrophil priming for an augmented respiratory burst, and induced loss of RBC deformability, compared with T/SS groups. Talactoferrin pretreatment significantly reduced the biological activity of T/HS lymph on respiratory burst activity and RBC deformability, but had no effect on the lymph cell count or endothelial cell permeability. CONCLUSIONS: These results provide a proof of principle that prophylactic dosing of oral talactoferrin can potentially protect the gut in a T/HS model and limit the production of biologically active factors in rat gastrointestinal tissue subjected to ischemia-reperfusion-type injuries.


Assuntos
Íleo/lesões , Lactoferrina/farmacologia , Linfa/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Choque Hemorrágico/tratamento farmacológico , Administração Oral , Animais , Íleo/efeitos dos fármacos , Laparotomia/efeitos adversos , Linfa/efeitos dos fármacos , Sistema Linfático/efeitos dos fármacos , Sistema Linfático/fisiologia , Masculino , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Traumatismo por Reperfusão/etiologia , Explosão Respiratória/efeitos dos fármacos , Choque Hemorrágico/etiologia , Ferimentos e Lesões/complicações
4.
Am J Physiol Gastrointest Liver Physiol ; 304(1): G57-63, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23125158

RESUMO

Recent studies demonstrate that mechanisms underlying gut barrier failure include systemic processes and less studied luminal processes. We thus tested the hypothesis that mucus layer oxidation is a component of trauma/hemorrhagic shock-induced gut injury and dysfunction. Male Sprague-Dawley rats underwent trauma/hemorrhagic shock. Controls underwent trauma only. Mucus from the terminal 30 cm of the ileum was collected, processed, and analyzed for reactive nitrogen intermediates (RNI)-mediated damage, reactive oxygen species (ROS)-induced damage, and total antioxidant capacity. The distal ileum was stained to quantify the mucus layer; gut permeability was assessed physiologically. A time course study was conducted to determine the temporal sequence of mucus layer damage. The role of free radical-mediated damage to the gut barrier was investigated by the effect of the free radical scavenger dimethyl sulfoxide on trauma/hemorrhagic shock-induced changes on the mucus and on gut permeability. Trauma/hemorrhagic shock increased intestinal permeability, which was associated with evidence of loss of the unstirred mucus layer. These changes correlated with increased ROS- and RNI-mediated mucus damage and loss of mucus total antioxidant capacity. Based on the time course study, ROS-mediated mucus damage and loss of total antioxidant capacity were present immediately following shock, whereas RNI-mediated damage was delayed for 3 h. Dimethyl sulfoxide ameliorated gut barrier loss, ROS-mediated changes to the mucus layer, and loss of total antioxidant capacity. There was no change in RNI-induced changes to the mucus layer. These results support the hypothesis that trauma/hemorrhagic shock leads to mucus damage and gut dysfunction through the generation of free radical species.


Assuntos
Mucosa Intestinal/metabolismo , Intestinos/lesões , Choque Hemorrágico/metabolismo , Animais , Antioxidantes/metabolismo , Dimetil Sulfóxido/farmacologia , Sequestradores de Radicais Livres/farmacologia , Mucosa Intestinal/fisiologia , Intestinos/fisiopatologia , Masculino , Oxidantes/metabolismo , Oxirredução , Permeabilidade , Carbonilação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Choque Hemorrágico/fisiopatologia , Tirosina/metabolismo
5.
J Surg Res ; 183(2): 678-85, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23465433

RESUMO

BACKGROUND: Myocardial contractile depression develops 4 to 24 h after major burn injury. We have reported previously that in a rat burn injury model (≈40% of total body surface area burn), mesenteric lymph duct ligation (LDL) prior to burn prevented myocardial dysfunction. However, the underlying cellular and molecular mechanisms are not well understood. MATERIALS AND METHODS: Left ventricular myocytes were isolated from sham burn (control), sham burn with LDL (sham + LDL), burn, and burn with LDL (burn + LDL) rats at 4 and 24 h after burn or sham burn. Electrophysiological techniques were used to study myocyte size, contractility and L-type Ca2+ channel current (ICa). Further studies examined changes in the messenger RNA expression levels of pore-forming subunit of the L-type Ca(2+) channel, α1C, and its auxiliary subunits, ß1, ß2, ß3, and α2δ1, which modulate the abundance of the ICa in post-burn hearts. RESULTS: Depressed myocyte contractility (≈20%) developed during 4 to 24 h post-burn compared with control, sham + LDL, or burn + LDL groups, a pattern of changes consistent with whole heart studies. There was no significant alteration in myocyte size. The ICa density was significantly decreased (≈30%) at 24 h post-burn, whereas the messenger RNA expression levels of Ca(2+) channel gene were not significantly altered at 4 and 24 h after burn injury. CONCLUSIONS: These results suggest that the post-burn contractile phenotype in vivo was also present in isolated myocytes in vitro, but cellular remodeling was not a major factor. The results also suggest that changes in ICa regulation, but not from Ca(2+) channel gene modification, may be a key element involved in post-burn contractile depression and the beneficial effects of LDL.


Assuntos
Queimaduras/complicações , Coração/fisiopatologia , Sistema Linfático/fisiopatologia , Mesentério/fisiopatologia , Contração Miocárdica/fisiologia , Miócitos Cardíacos/patologia , Animais , Canais de Cálcio Tipo L/fisiologia , Tamanho Celular , Técnicas In Vitro , Ligadura , Masculino , Modelos Animais , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Fatores de Tempo
6.
J Immunol ; 186(7): 4340-6, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21339364

RESUMO

Classically, sympathetic and parasympathetic systems act in opposition to maintain the physiological homeostasis. In this article, we report that both systems work together to restrain systemic inflammation in life-threatening conditions such as sepsis. This study indicates that vagus nerve and cholinergic agonists activate the sympathetic noradrenergic splenic nerve to control systemic inflammation. Unlike adrenalectomy, splenectomy and splenic neurectomy prevent the anti-inflammatory potential of both the vagus nerve and cholinergic agonists, and abrogate their potential to induce splenic and plasma norepinephrine. Splenic nerve stimulation mimics vagal and cholinergic induction of norepinephrine and re-establishes neuromodulation in α7 nicotinic acetylcholine receptor (α7nAChR)-deficient animals. Thus, vagus nerve and cholinergic agonists inhibit systemic inflammation by activating the noradrenergic splenic nerve via the α7nAChR nicotinic receptors. α7nAChR represents a unique molecular link between the parasympathetic and sympathetic system to control inflammation.


Assuntos
Norepinefrina/biossíntese , Receptores Nicotínicos/fisiologia , Baço/imunologia , Baço/metabolismo , Estimulação do Nervo Vago/métodos , Animais , Agonistas Colinérgicos/farmacologia , Vias Eferentes/imunologia , Vias Eferentes/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Norepinefrina/sangue , Sistema Nervoso Parassimpático/imunologia , Sistema Nervoso Parassimpático/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/deficiência , Receptores Nicotínicos/genética , Baço/inervação , Sistema Nervoso Simpático/imunologia , Sistema Nervoso Simpático/metabolismo , Receptor Nicotínico de Acetilcolina alfa7
7.
J Immunol ; 187(2): 718-25, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21666060

RESUMO

Many anti-inflammatory strategies that are successful in treating sepsis in healthy animals fail in clinical trials, in part because sepsis normally involves immunocompromised patients, and massive lymphocyte apoptosis prevents immunomodulation. In this article, we report a new set of regulatory lymphocytes that are able to re-establish the cholinergic anti-inflammatory modulation and to provide therapeutic advantages in sepsis. The vagus nerve controls inflammation in healthy, but not in septic, mice. Likewise, vagus nerve and cholinergic agonists fail to control inflammation in splenectomized and nude animals. Unlike typical suppressor CD25(+) cells, CD4(+)CD25(-) lymphocytes re-establish the anti-inflammatory potential of the vagus nerve and cholinergic agonists in immunocompromised and septic animals. These cholinergic lymphocytes re-establish splenic protection and the potential of cholinergic agonists to rescue immunocompromised animals from established sepsis. The study results revealed these new regulatory lymphocytes as, to our knowledge, the first known physiological target for neuromodulation of the innate immune responses and a potential therapeutic target for sepsis.


Assuntos
Imunidade Inata , Neuroimunomodulação/imunologia , Receptores Nicotínicos/fisiologia , Sepse/imunologia , Sepse/fisiopatologia , Linfócitos T Reguladores/imunologia , Nervo Vago/fisiologia , Animais , Ceco , Endotoxemia/imunologia , Endotoxemia/mortalidade , Endotoxemia/prevenção & controle , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Nus , Punções , Distribuição Aleatória , Sepse/mortalidade , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Receptor Nicotínico de Acetilcolina alfa7
8.
J Immunol ; 186(11): 6465-73, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21502376

RESUMO

Cellular X-chromosome mosaicism, which is unique to females, may be advantageous during pathophysiological challenges compared with the single X-chromosome machinery of males, and it may contribute to gender dimorphism in the inflammatory response. We tested the hypothesis of whether cellular mosaicism for the X-linked gp91phox (NOX2) deficiency, the catalytic component of the superoxide anion-generating NADPH oxidase complex, is advantageous during polymicrobial sepsis. Deficient, wild-type (WT), and heterozygous/mosaic mice were compared following polymicrobial sepsis initiated by cecal ligation and puncture. Compared with WT littermates, sepsis-induced mortality was improved in deficient mice, as well as in mosaic animals carrying both deficient and WT phagocyte subpopulations. In contrast, blood bacterial counts were greatest in deficient mice. Consistent with poor survival, WT mice also showed the most severe organ damage following sepsis. In mosaic animals, the deficient neutrophil subpopulations displayed increased organ recruitment and elevated CD11b membrane expression compared with WT neutrophil subpopulations within the same animal. The dynamics of sepsis-induced blood and organ cytokine content and WBC composition changes, including lymphocyte subsets in blood and bone marrow, showed differences among WT, deficient, and mosaic subjects, indicating that mosaic mice are not simply the average of the deficient and WT responses. Upon oxidative burst, interchange of oxidants between WT and deficient neutrophil subpopulations occurred in mosaic mice. This study suggests that mice mosaic for gp91phox expression have multiple advantages compared with WT and deficient mice during the septic course.


Assuntos
Bacteriemia/genética , Inflamação/genética , Glicoproteínas de Membrana/genética , NADPH Oxidases/genética , Animais , Bacteriemia/metabolismo , Bacteriemia/microbiologia , Carga Bacteriana , Células da Medula Óssea/metabolismo , Antígeno CD11b/sangue , Ceco/cirurgia , Citocinas/sangue , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Inflamação/metabolismo , Inflamação/patologia , Contagem de Leucócitos , Ligadura , Masculino , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mosaicismo , NADPH Oxidase 2 , NADPH Oxidases/deficiência , Neutrófilos/metabolismo , Punções , Cromossomo X/genética
9.
J Immunol ; 187(8): 4256-67, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21918191

RESUMO

The extracellular concentrations of adenosine are increased during sepsis, and adenosine receptors regulate the host's response to sepsis. In this study, we investigated the role of the adenosine-generating ectoenzyme, ecto-5'-nucleotidase (CD73), in regulating immune and organ function during sepsis. Polymicrobial sepsis was induced by subjecting CD73 knockout (KO) and wild type (WT) mice to cecal ligation and puncture. CD73 KO mice showed increased mortality in comparison with WT mice, which was associated with increased bacterial counts and elevated inflammatory cytokine and chemokine concentrations in the blood and peritoneum. CD73 deficiency promoted lung injury, as indicated by increased myeloperoxidase activity and neutrophil infiltration, and elevated pulmonary cytokine levels. CD73 KO mice had increased apoptosis in the thymus, as evidenced by increased cleavage of caspase-3 and poly(ADP-ribose) polymerase and increased activation of NF-κB. Septic CD73 KO mice had higher blood urea nitrogen levels and increased cytokine levels in the kidney, indicating increased renal dysfunction. The increased kidney injury of CD73 KO mice was associated with augmented activation of p38 MAPK and decreased phosphorylation of Akt. Pharmacological inactivation of CD73 in WT mice using α, ß-methylene ADP augmented cytokine levels in the blood and peritoneal lavage fluid. These findings suggest that CD73-derived adenosine may be beneficial in sepsis.


Assuntos
5'-Nucleotidase/metabolismo , Sepse/metabolismo , Sepse/fisiopatologia , 5'-Nucleotidase/imunologia , Adenosina/imunologia , Adenosina/metabolismo , Animais , Western Blotting , Separação Celular , Quimiocinas/análise , Quimiocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Knockout , Sepse/imunologia
10.
Am J Physiol Gastrointest Liver Physiol ; 303(8): G969-78, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22899820

RESUMO

Recent studies have shown that mesenteric lymph plays a very important role in the development of multiple-organ dysfunction syndrome under critical conditions. Great efforts have been made to identify the biologically active molecules in the lymph. We used a trauma-hemorrhagic shock (T/HS) model and the superior mesenteric artery occlusion (SMAO) model, representing a global and a localized intestinal ischemia-reperfusion insult, respectively, to investigate the role of free fatty acids (FFAs) in the cytotoxicity of mesenteric lymph in rats. Lymph was collected before, during, and after (post) shock or SMAO. The post-T/HS and SMAO lymph, but not the sham lymph, manifested cytotoxicity for human umbilical vein endothelial cells (HUVECs). HUVEC cytotoxicity was associated with increased FFAs, especially the FFA-to-protein ratio. Addition of albumin, especially delipidated albumin, reduced this cytotoxicity. Lipase treatment of trauma-sham shock (T/SS) lymph converted it from a noncytotoxic to a cytotoxic fluid, and its toxicity correlated with the FFA-to-protein ratio in a fashion similar to that of the T/HS lymph, further suggesting that FFAs were the key components leading to HUVEC cytotoxicity. Analysis of lymph by gas chromatography revealed that the main FFAs in the post-T/HS or lipase-treated T/SS lymph were palmitic, stearic, oleic, and linoleic acids. When added to the cell culture at levels comparable to those in T/HS lymph, all these FFAs were cytotoxic, with linoleic acid being the most potent. In conclusion, this study suggests that lipase-generated FFAs are the key components resulting in the cytotoxicity of T/HS and SMAO mesenteric lymph.


Assuntos
Ácidos Graxos não Esterificados/análise , Lipase/análise , Linfa/química , Oclusão Vascular Mesentérica/fisiopatologia , Choque Hemorrágico/fisiopatologia , Animais , Endotélio Vascular/fisiopatologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Artéria Mesentérica Superior/fisiopatologia , Ratos , Ratos Sprague-Dawley
11.
FASEB J ; 25(12): 4476-85, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21840939

RESUMO

The nervous system is classically organized into sympathetic and parasympathetic systems acting in opposition to maintain physiological homeostasis. Here, we report that both systems converge in the activation of ß2-adrenoceptors of splenic regulatory lymphocytes to control systemic inflammation. Vagus nerve stimulation fails to control serum TNF levels in either ß2-knockout or lymphocyte-deficient nude mice. Unlike typical suppressor CD25(+) cells, the transfer of CD4(+)CD25(-) regulatory lymphocytes reestablishes the anti-inflammatory potential of the vagus nerve and ß2-agonists to control inflammation in both ß2-knockout and nude mice. ß2-Agonists inhibit cytokine production in splenocytes (IC(50)≈ 1 µM) and prevent systemic inflammation in wild-type but not in ß2-knockout mice. ß2-Agonists rescue wild-type mice from established polymicrobial peritonitis in a clinically relevant time frame. Regulatory lymphocytes reestablish the anti-inflammatory potential of ß2-agonists to control systemic inflammation, organ damage, and lethal endotoxic shock in ß2-knockout mice. These results indicate that ß2-adrenoceptors in regulatory lymphocytes are critical for the anti-inflammatory potential of the parasympathetic vagus nerve, and they represent a potential pharmacological target for sepsis.


Assuntos
Imunidade Inata , Neuroimunomodulação , Receptores Adrenérgicos beta 2/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Citocinas/biossíntese , Endotoxemia/imunologia , Endotoxemia/fisiopatologia , Hemodinâmica , Inflamação/imunologia , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Norepinefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta 2/deficiência , Receptores Adrenérgicos beta 2/genética , Sepse/imunologia , Sepse/fisiopatologia , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue , Nervo Vago/imunologia
12.
J Immunol ; 185(1): 542-50, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20505145

RESUMO

Despite intensive research, efforts to reduce the mortality of septic patients have failed. Adenosine is a potent extracellular signaling molecule, and its levels are elevated in sepsis. Adenosine signals through G-protein-coupled receptors and can regulate the host's response to sepsis. In this study, we studied the role of A(2B) adenosine receptors in regulating the mortality and inflammatory response of mice following polymicrobial sepsis. Genetic deficiency of A(2B) receptors increased the mortality of mice suffering from cecal ligation and puncture-induced sepsis. The increased mortality of A(2B) knockout mice was associated with increased levels of inflammatory cytokines and chemokines and augmented NF-kappaB and p38 activation in the spleen, heart, and plasma in comparison with wild-type animals. In addition, A(2B) receptor knockout mice showed increased splenic apoptosis and phosphatase and tensin homolog activation and decreased Akt activation. Experiments using bone-marrow chimeras revealed that it is the lack of A(2B) receptors on nonhematopoietic cells that is primarily responsible for the increased inflammation of septic A(2B) receptor-deficient mice. These results indicate that A(2B) receptor activation may offer a new therapeutic approach for the management of sepsis.


Assuntos
Mediadores da Inflamação/fisiologia , Receptor A2B de Adenosina/fisiologia , Sepse/mortalidade , Sepse/prevenção & controle , Lesão Pulmonar Aguda/microbiologia , Lesão Pulmonar Aguda/mortalidade , Lesão Pulmonar Aguda/prevenção & controle , Antagonistas do Receptor A2 de Adenosina , Animais , Ceco , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/microbiologia , Peritonite/mortalidade , Peritonite/prevenção & controle , Punções/efeitos adversos , Receptor A2B de Adenosina/deficiência , Sepse/microbiologia , Taxa de Sobrevida
13.
Anesth Analg ; 115(1): 118-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22584546

RESUMO

BACKGROUND: Mechanical ventilation (MV) can lead to ventilator-induced lung injury secondary to trauma and associated increases in pulmonary inflammatory cytokines. There is controversy regarding the associated systemic inflammatory response. In this report, we demonstrate the effects of MV on systemic inflammation. METHODS: This report is part of a previously published study (Hong et al. Anesth Analg 2010;110:1652-60). Female pigs were randomized into 3 groups. Group H-Vt/3 was ventilated with a tidal volume (Vt) of 15 mL/kg predicted body weight (PBW)/positive end-expiratory pressure (PEEP) of 3 cm H(2)O; group L-Vt/3 with a Vt of 6 mL/kg PBW/PEEP of 3 cm H2O; and group L-Vt/10 with a Vt of 6 mL/kg PBW/PEEP of 10 cm H(2)O, for 8 hours. Each group had 6 subjects (n = 6). Prelung and postlung sera were analyzed for inflammatory markers. Hemodynamics, airway mechanics, and arterial blood gases were monitored. RESULTS: There were no significant differences in systemic cytokines among groups. There were similar trends of serum inflammatory markers in all subjects. This is in contrast to findings previously published demonstrating increases in inflammatory mediators in bronchoalveolar lavage. CONCLUSION: Systemic inflammatory markers did not correlate with lung injury associated with MV.


Assuntos
Lesão Pulmonar Aguda/etiologia , Respiração com Pressão Positiva/efeitos adversos , Síndrome de Resposta Inflamatória Sistêmica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/fisiopatologia , Animais , Citocinas/sangue , Feminino , Hemodinâmica , Mediadores da Inflamação/sangue , Suínos , Síndrome de Resposta Inflamatória Sistêmica/sangue , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Volume de Ventilação Pulmonar , Fatores de Tempo , Lesão Pulmonar Induzida por Ventilação Mecânica/sangue , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia
14.
Surgeon ; 10(6): 350-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22534256

RESUMO

The concept of bacterial translocation and gut-origin sepsis as a cause of systemic infectious complications and the multiple organ dysfunction syndrome (MODS) in surgical and ICU patients has emerged over the last several decades, although the exact clinical relevance of these phenomena continues to be debated. Thus, the goal of this review is to trace the evolution of gut-origin sepsis and gut-induced MODS and put these disorders and observations into clinical perspective. Additionally, the mechanisms leading to gut-derived complications are explored as well as therapeutic options to limit or prevent these complications. From this work, several major conclusions emerge. First, that bacterial translocation occurs clinically and is responsible for increased infectious complications in patients undergoing major abdominal surgery. However, the phenomenon of bacterial translocation is not sufficient to explain the development of MODS in ICU patients. Instead, the development of MODS in these high-risk patients is likely due to gut injury and the systemic spread of non-microbial, tissue-injurious factors that reach the systemic circulation via the intestinal lymphatics. These observations have resulted in the gut-lymph hypothesis of MODS.


Assuntos
Bactérias/isolamento & purificação , Fenômenos Fisiológicos Bacterianos , Translocação Bacteriana , Intestinos/microbiologia , Sepse/microbiologia , Humanos
15.
Am J Physiol Gastrointest Liver Physiol ; 300(5): G853-61, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21183660

RESUMO

Gut injury and loss of normal intestinal barrier function are key elements in the paradigm of gut-origin systemic inflammatory response syndrome, acute lung injury, and multiple organ dysfunction syndrome (MODS). As hypoxia-inducible factor (HIF-1) is a critical determinant of the physiological and pathophysiological response to hypoxia and ischemia, we asked whether HIF-1 plays a proximal role in the induction of gut injury and subsequent lung injury. Using partially HIF-1α-deficient mice in an isolated superior mesenteric artery occlusion (SMAO) intestinal ischemia reperfusion (I/R) injury model (45 min SMAO followed by 3 h of reperfusion), we showed a direct relationship between HIF-1 activation and intestinal I/R injury. Specifically, partial HIF-1α deficiency attenuated SMAO-induced increases in intestinal permeability, lipid peroxidation, mucosal caspase-3 activity, and IL-1ß mRNA levels. Furthermore, partial HIF-1α deficiency prevented the induction of ileal mucosal inducible nitric oxide synthase (iNOS) protein levels after SMAO and iNOS deficiency ameliorated SMAO-induced villus injury. Resistance to SMAO-induced gut injury was also associated with resistance to lung injury, as reflected by decreased levels of myeloperoxidase, IL-6 and IL-10 in the lungs of HIF-1α(+/-) mice. In contrast, a short duration of SMAO (15 min) followed by 3 h of reperfusion neither induced mucosal HIF-1α protein levels nor caused significant gut and lung injury in wild-type or HIF-1α(+/-) mice. This study indicates that intestinal HIF-1 activation is a proximal regulator of I/R-induced gut mucosal injury and gut-induced lung injury. However, the duration and severity of the gut I/R insult dictate whether HIF-1 plays a gut-protective or deleterious role.


Assuntos
Fator 1 Induzível por Hipóxia/fisiologia , Enteropatias/patologia , Traumatismo por Reperfusão/patologia , Lesão Pulmonar Aguda/patologia , Animais , Western Blotting , Caspase 3/metabolismo , Ensaio de Imunoadsorção Enzimática , Genótipo , Fator 1 Induzível por Hipóxia/genética , Mucosa Intestinal/fisiologia , Intestinos/irrigação sanguínea , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Permeabilidade , Peroxidase/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Eur J Immunol ; 40(9): 2580-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20706987

RESUMO

The role of STAT3 in infectious diseases remains undetermined, in part because unphosphorylated STAT3 has been considered an inactive protein. Here, we report that unphosphorylated STAT3 contributes to cholinergic anti-inflammation, prevents systemic inflammation, and improves survival in sepsis. Bacterial endotoxin induced STAT3 tyrosine phosphorylation in macrophages. Both alpha 7 nicotinic receptor (alpha 7nAChR) activation and inhibition of JAK2 blunt STAT3 phosphorylation. Inhibition of STAT3 phosphorylation mimicked the alpha 7nAChR signaling, inhibiting NF-kappaB and cytokine production in macrophages. Transfection of macrophages with the dominant-negative mutant STAT3F, to prevent its tyrosine phosphorylation, reduced TNF production but did not prevent the alpha 7nAChR signaling. However, inhibition of STAT3 protein expression enhanced cytokine production and abrogated alpha 7nAChR signaling. Alpha 7nAChR controls TNF production in macrophages through a mechanism that requires STAT3 protein expression, but not its tyrosine phosphorylation. In vivo, inhibition of STAT3 tyrosine phosphorylation by stattic prevented systemic inflammation and improved survival in experimental sepsis. Stattic also prevented the production of late mediators of sepsis and improved survival in established sepsis. These results reveal the immunological implications of tyrosine-unphosphorylated STAT3 in infectious diseases.


Assuntos
Macrófagos/metabolismo , Proteínas Mutantes/metabolismo , Receptores Nicotínicos/metabolismo , Fator de Transcrição STAT3/metabolismo , Sepse/imunologia , Animais , Anti-Inflamatórios/administração & dosagem , Linhagem Celular , Óxidos S-Cíclicos/administração & dosagem , Imunomodulação , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mutantes/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , RNA Interferente Pequeno/genética , Receptores Nicotínicos/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Sepse/tratamento farmacológico , Sepse/genética , Sepse/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transgenes/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Receptor Nicotínico de Acetilcolina alfa7
17.
FASEB J ; 24(8): 2631-40, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20371613

RESUMO

Activation-induced cell death (AICD) is initiated by T-cell receptor (TCR) restimulation of already activated and expanded peripheral T cells and is mediated through Fas/Fas ligand (FasL) interactions. Adenosine is a purine nucleoside signaling molecule, and its immunomodulatory effects are mediated by 4 G-protein-coupled receptors: A(1), A(2A), A(2B), and A(3). In this study, we investigated the role of A(2A) receptors in regulating CD4(+) T lymphocyte AICD. Our results showed that the selective A(2A) receptor agonist CGS21680 (EC(50)=15.2-32.6 nM) rescued mouse CD4(+) hybridomas and human Jurkat cells from AICD and that this effect was reversed by the selective A(2A) receptor antagonist ZM241385 (EC(50)=2.3 nM). CGS21680 decreased phosphatidylserine exposure on the membrane, as well as the cleavage of caspase-3, caspase-8 and poly(ADP-ribose) polymerase indicating that A(2A) receptor stimulation blocks the extrinsic apoptotic pathway. In addition, CGS21680 attenuated both Fas and FasL mRNA expression. This decrease in FasL expression was associated with decreased activation of the transcription factor systems NF-kappaB, NF-ATp, early growth response (Egr)-1, and Egr-3. The antiapoptotic effect of A(2A) receptor stimulation was mediated by protein kinase A. Together, these results demonstrate that A(2A) receptor activation suppresses the AICD of peripheral T cells.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/citologia , Ativação Linfocitária , Receptor A2A de Adenosina/metabolismo , Animais , Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Morte Celular , Proteína Ligante Fas/genética , Humanos , Células Jurkat , Camundongos , Substâncias Protetoras , Receptor fas/genética
18.
Circ Res ; 105(2): 158-66, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19542014

RESUMO

Phenotypic modulation of vascular smooth muscle cells (VSMCs) plays a critical role in the pathogenesis of a variety of proliferative vascular diseases. Recently, we have found that microRNA (miRNA) miR-145 is the most abundant miRNA in normal vascular walls and in freshly isolated VSMCs; however, the role of miR-145 in VSMC phenotypic modulation and vascular diseases is currently unknown. Here we find that miR-145 is selectively expressed in VSMCs of the vascular wall and its expression is significantly downregulated in the vascular walls with neointimal lesion formation and in cultured dedifferentiated VSMCs. More importantly, both in cultured rat VSMCs in vitro and in balloon-injured rat carotid arteries in vivo, we demonstrate that the noncoding RNA miR-145 is a novel phenotypic marker and a novel phenotypic modulator of VSMCs. VSMC differentiation marker genes such as SM alpha-actin, calponin, and SM-MHC are upregulated by premiR-145 or adenovirus expressing miR-145 (Ad-miR-145) but are downregulated by the miR-145 inhibitor 2'OMe-miR-145. We have further identified that miR-145-mediated phenotypic modulation of VSMCs is through its target gene KLF5 and its downstream signaling molecule, myocardin. Finally, restoration of miR-145 in balloon-injured arteries via Ad-miR-145 inhibits neointimal growth. We conclude that miR-145 is a novel VSMC phenotypic marker and modulator that is able of controlling vascular neointimal lesion formation. These novel findings may have extensive implications for the diagnosis and therapy of a variety of proliferative vascular diseases.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Proliferação de Células , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Túnica Íntima/metabolismo , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Cateterismo/efeitos adversos , Desdiferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Hiperplasia , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , MicroRNAs/genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Proteínas Nucleares/metabolismo , Fenótipo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transativadores/metabolismo , Transfecção , Túnica Íntima/patologia
19.
J Surg Res ; 169(2): 257-66, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20189589

RESUMO

BACKGROUND: Alternative experimental models of hemorrhage mimic particular conditions of clinical settings and provide advantages to analyze novel resuscitation treatments. Here, we compared alternative models of hemorrhage and analyzed the effects of resuscitation with Hextend. METHODS: Adult male Sprague-Dawley rats underwent alternative models of hemorrhage: anesthetized without trauma, anesthetized with trauma, or conscious (unanesthetized) hemorrhage. Each model of hemorrhage includes three experimental groups: (C) control without hemorrhage or resuscitation treatment; (NR) animals with hemorrhage but without resuscitation; and (HX) animals with hemorrhage and resuscitation treatment with Hextend. RESULTS: Conscious animals required the highest hemorrhagic volume, whereas hemorrhage with trauma required the lowest blood volume withdrawal to achieve the same arterial pressure. Conscious hemorrhage exhibited the fastest mortality, but anesthetized animals with or without trauma had similar mortality kinetic. These survival rates did not correlate with blood chemistry, hemodynamic responses, or serum TNF and HMGB1 levels. Hemorrhage in conscious animals or anesthetized animals with trauma increased serum TNF levels by approximately 2-fold compared with hemorrhage in anesthetized animals without trauma. Animals in conscious hemorrhage had similar TNF increases in all the organs, but trauma induced a specific TNF overproduction in the spleen. Resuscitation with Hextend improved survival in all the experimental models, yet its survival benefits were statistically greater in anesthetized animals with trauma. The only two markers similar to the survival benefits of Hextend were the TNF levels in the lung and liver. Hextend significantly improved survival and inhibited pulmonary and hepatic TNF levels in all the experimental models. CONCLUSIONS: The survival benefits of resuscitation with Hextend depended on the experimental models and did not correlate with blood chemistry, hemodynamic, or serum cytokine levels. However, resuscitation with Hextend inhibited TNF levels in the lung and the liver with a pattern that resembled the survival benefits.


Assuntos
Hemorragia/fisiopatologia , Inflamação/fisiopatologia , Modelos Animais , Sobrevida/fisiologia , Inconsciência , Ferimentos e Lesões , Animais , Proteína HMGB1/metabolismo , Hemorragia/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Substitutos do Plasma/uso terapêutico , Ratos , Ratos Sprague-Dawley , Ressuscitação/métodos , Fator de Necrose Tumoral alfa/metabolismo
20.
J Surg Res ; 166(2): e135-42, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21276979

RESUMO

BACKGROUND: Plasma factor XIII (FXIII) is responsible for stabilization of fibrin clot at the final stage of blood coagulation. Since FXIII has also been shown to modulate inflammation, endothelial permeability, as well as diminish multiple organ dysfunction (MOD) after gut ischemia-reperfusion injury, we hypothesized that FXIII would reduce MOD caused by trauma-hemorrhagic shock (THS). MATERIALS AND METHODS: Rats were subjected to a 90 min THS or trauma sham shock (TSS) and treated with either recombinant human FXIII A(2) subunit (rFXIII) or placebo immediately after resuscitation with shed blood or at the end of the TSS period. Lung permeability, lung and gut myeloperoxidase (MPO) activity, gut histology, neutrophil respiratory burst, microvascular blood flow in the liver and muscles, and cytokine levels were measured 3 h after the THS or TSS. FXIII levels were measured before THS or TSS and after the 3-h post-shock period. RESULTS: THS-induced lung permeability as well as lung and gut MPO activity was significantly lower in rFXIII-treated than in placebo-treated animals. Similarly, rFXIII-treated rats had lower neutrophil respiratory burst activity and less ileal mucosal injury. rFXIII-treated rats also had a higher liver microvascular blood flow compared with the placebo group. Cytokine response was more favorable in rFXIII-treated animals. Trauma-hemorrhagic shock did not cause a drop in FXIII activity during the study period. CONCLUSIONS: Administration of rFXIII diminishes THS-induced MOD in rats, presumably by preservation of the gut barrier function, limitation of polymorphonuclear leukocyte (PMN) activation, and modulation of the cytokine response.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Fator XIII/farmacologia , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Choque Hemorrágico/tratamento farmacológico , Lesão Pulmonar Aguda/etiologia , Animais , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Humanos , Íleo/irrigação sanguínea , Fígado/irrigação sanguínea , Pulmão/irrigação sanguínea , Masculino , Microcirculação/efeitos dos fármacos , Insuficiência de Múltiplos Órgãos/etiologia , Neutrófilos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Explosão Respiratória/efeitos dos fármacos , Choque Hemorrágico/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA