Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(5)2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38473750

RESUMEN

Uncontrolled hemorrhage is a major preventable cause of death in patients with trauma. However, the majority of large animal models of hemorrhage have utilized controlled hemorrhage rather than uncontrolled hemorrhage to investigate the impact of immunopathy and coagulopathy on multi-organ failure (MOF) and mortality. This study evaluates these alterations in a severe porcine controlled and uncontrolled hemorrhagic shock (HS) model. Anesthetized female swine underwent controlled hemorrhage and uncontrolled hemorrhage by partial splenic resection followed with or without lactated Ringer solution (LR) or Voluven® resuscitation. Swine were surveyed 6 h after completion of splenic hemorrhage or until death. Blood chemistry, physiologic variables, systemic and tissue levels of complement proteins and cytokines, coagulation parameters, organ function, and damage were recorded and assessed. HS resulted in systemic and local complement activation, cytokine release, hypocoagulopathy, metabolic acidosis, MOF, and no animal survival. Resuscitation with LR and Voluven® after HS improved hemodynamic parameters (MAP and SI), metabolic acidosis, hyperkalemia, and survival but resulted in increased complement activation and worse coagulopathy. Compared with the LR group, the animals with hemorrhagic shock treated with Voluven® had worse dilutional anemia, coagulopathy, renal and hepatic dysfunction, increased myocardial complement activation and renal damage, and decreased survival rate. Hemorrhagic shock triggers early immunopathy and coagulopathy and appears associated with MOF and death. This study indicates that immunopathy and coagulopathy are therapeutic targets that may be addressed with a high-impact adjunctive treatment to conventional resuscitation.


Asunto(s)
Acidosis , Trastornos de la Coagulación Sanguínea , Choque Hemorrágico , Humanos , Femenino , Porcinos , Animales , Insuficiencia Multiorgánica , Hemorragia , Citocinas
2.
J Immunol ; 198(2): 788-797, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27913632

RESUMEN

Intestinal ischemia followed by reperfusion leads to local and remote organ injury attributed to inflammatory response during the reperfusion phase. The extent to which ischemia contributes to ischemia/reperfusion injury has not been thoroughly studied. After careful evaluation of intestinal tissue following 30 min of ischemia, we noticed significant local mucosal injury in wild-type mice. This injury was drastically reduced in C3-deficient mice, suggesting C3 involvement. Depletion of circulating complement with cobra venom factor eliminated, as expected, injury recorded at the end of the reperfusion phase but failed to eliminate injury that occurred during the ischemic phase. Immunohistochemical studies showed that tissue damage during ischemia was associated with increased expression of C3/C3 fragments primarily in the intestinal epithelial cells, suggesting local involvement of complement. In vitro studies using Caco2 intestinal epithelial cells showed that in the presence of LPS or exposure to hypoxic conditions the cells produce higher C3 mRNA as well as C3a fragment. Caco2 cells were also noted to produce cathepsins B and L, and inhibition of cathepsins suppressed the release of C3a. Finally, we found that mice treated with a cathepsin inhibitor and cathepsin B-deficient mice suffer limited intestinal injury during the ischemic phase. To our knowledge, our findings demonstrate for the first time that significant intestinal injury occurs during ischemia prior to reperfusion and that this is due to activation of C3 within the intestinal epithelial cells in a cathepsin-dependent manner. Modulation of cathepsin activity may prevent injury of organs exposed to ischemia.


Asunto(s)
Complemento C3/metabolismo , Isquemia Mesentérica/metabolismo , Daño por Reperfusión/metabolismo , Animales , Western Blotting , Células CACO-2 , Catepsinas/metabolismo , Complemento C3/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Isquemia Mesentérica/inmunología , Isquemia Mesentérica/patología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología
3.
J Immunol ; 199(8): 2921-2929, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28877988

RESUMEN

Ischemia-reperfusion (IR) injury to the small intestine following clamping of the superior mesenteric artery results in an intense local inflammatory response that is characterized by villous damage and neutrophil infiltration. IL-17A, a cytokine produced by a variety of cells in response to inflammatory cytokines released following tissue injury, has been implicated in IR injury. Using Il17a-/- , Il23r-/- , and Rorc-/- mice and administration of anti-IL-17A and anti-IL-23 neutralizing Abs to wild-type mice, we demonstrate that intestinal IR injury depends on IL-17A and that IL-17A is downstream of the binding of autoantibody to ischemia-conditioned tissues and subsequent complement activation. Using bone marrow chimeras, we demonstrate that the IL-17A required for intestinal IR injury is derived from hematopoietic cells. Finally, by transferring autoantibody-rich sera into Rag2γc-/- and Rag2-/- mice, we demonstrate that innate lymphoid cells are the main producers of IL-17A in intestinal IR injury. We propose that local production of IL-17A by innate lymphoid cells is crucial for the development of intestinal IR injury and may provide a therapeutic target for clinical exploitation.


Asunto(s)
Interleucina-17/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/patología , Linfocitos/inmunología , Daño por Reperfusión/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Autoanticuerpos/metabolismo , Células Cultivadas , Activación de Complemento , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Interleucina-17/genética , Arteria Mesentérica Superior/cirugía , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Receptores de Interleucina/genética
4.
Proc Natl Acad Sci U S A ; 110(35): 14348-53, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23942120

RESUMEN

Inflammation and vascular injury triggered by ischemia/reperfusion (I/R) represent a leading cause of morbidity and mortality in a number of clinical settings. Wnt and its homolog partners R-spondins, in addition to regulating embryonic development have recently been demonstrated to serve as wound-healing agents in inflammation-associated conditions. Here we ask whether R-spondins could prevent inflammation-associated tissue damage in ischemic disorders and thus investigate the role of R-spondin3 (R-spo3) in a mouse model of mesenteric I/R. We demonstrate that R-spo3 ameliorates mesenteric I/R-induced local intestinal as well as remote lung damage by suppressing local and systemic cytokine response and deposition of IgM and complement in intestinal tissues. We also show that decreased inflammatory response is accompanied by tightening of endothelial cell junctions and reduction in vascular leakage. We conclude that R-spo3 stabilizes endothelial junctions and inhibits vascular leakage during I/R and thereby mitigates the inflammatory events and associated tissue damage. Our findings uniquely demonstrate a protective effect of R-spo3 in I/R-related tissue injury and suggest a mechanism by which it may have these effects.


Asunto(s)
Endotelio Vascular/metabolismo , Isquemia Mesentérica/metabolismo , Trombospondinas/metabolismo , Animales , Endotelio Vascular/patología , Uniones Intercelulares/efectos de los fármacos , Isquemia Mesentérica/tratamiento farmacológico , Ratones , Unión Proteica , Daño por Reperfusión/prevención & control , Trombospondinas/farmacología
5.
Glycobiology ; 24(4): 379-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24440830

RESUMEN

ß-Glucans possess broad immunomodulatory properties, including activation of innate immune functions such as oxidative burst activity. The differential roles of complement receptor type 3 (CR3) and Dectin-1, the known ß-glucan receptors, and their associated signaling pathways in the generation of oxidative burst induced by different physical forms of Saccharomyces cerevisiae-derived ß-glucan were examined in human peripheral blood mononuclear cells (PBMC). In this study whole glucan particle (WGP) or immobilized soluble ß-glucan (ISG) was used to represent the phagocytizable or the nonphagocytizable form of a fungus, respectively. Oxidative burst as measured by the formation of superoxide (SO) was detected in PBMC in response to WGP and ISG. SO induction with WGP was concluded to be Dectin-1-mediated and required Src family kinases, phosphatidylinositol-3 kinase and protein kinase B/Akt. In contrast, the SO induction generated by ISG was CR3-mediated and required focal adhesion kinase, spleen tyrosine kinase, phosphatidylinositol-3 kinase, Akt, p38 mitogen activated protein kinase, phospholipase C and protein kinase C. The study results support the hypothesis that human PBMC, specifically monocytes, utilize distinct receptors and overlapping, but distinct, signaling pathways for the oxidative burst in response to challenge by different physical forms of ß-glucan.


Asunto(s)
Lectinas Tipo C/metabolismo , Leucocitos Mononucleares/metabolismo , Receptores de Complemento/metabolismo , Estallido Respiratorio , Transducción de Señal , Humanos
6.
Crit Care Med ; 42(5): e364-72, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24448198

RESUMEN

OBJECTIVE: Complement system is activated in patients with trauma. Although complement activation is presumed to contribute to organ damage and constitutional symptoms, little is known about the involved mechanisms. Because complement components may deposit on RBCs, we asked whether complement deposits on the surface of RBC in trauma and whether such deposition alters RBC function. DESIGN: A prospective experimental study. SETTING: Research laboratory. SUBJECTS: Blood samples collected from 42 trauma patients and 21 healthy donors. INTERVENTION: None. MEASUREMENTS AND MAIN RESULTS: RBC and sera were collected from trauma patients and control donors. RBCs from trauma patients (n = 40) were found to display significantly higher amounts of C4d on their surface by flow cytometry compared with RBCs from control (n = 17) (p < 0.01). Increased amounts of iC3b were found in trauma sera (n = 27) (vs 12 controls, p < 0.01) by enzyme-linked immunosorbent assay. Incubation of RBC from universal donors (type O, Rh negative) with trauma sera (n = 10) promoted C4d deposition on their surface (vs six controls, p< 0.05). Complement-decorated RBC (n = 6) displayed limited their deformability (vs six controls, p < 0.05) in two-dimensional microchannel arrays. Incubation of RBC with trauma sera (n = 10) promoted the phosphorylation of band 3, a cytoskeletal protein important for the function of the RBC membrane (vs eight controls, p < 0.05), and also accelerated calcium influx (n = 9) and enhanced nitric oxide production (n = 12) (vs four and eight controls respectively, p < 0.05) in flow cytometry. CONCLUSIONS: Our study found the presence of extensive complement activation in trauma patients and presents new evidence in support of the hypothesis that complement activation products deposit on the surface of RBC. Such deposition could limit RBC deformability and promote the production of nitric oxide. Our findings suggest that RBC in trauma patients malfunctions, which may explain organ damage and constitutional symptoms that is not accounted for otherwise by previously known pathophysiologic mechanisms.


Asunto(s)
Calcio/sangre , Activación de Complemento/fisiología , Eritrocitos/metabolismo , Óxido Nítrico/sangre , Fragmentos de Péptidos/sangre , Heridas y Lesiones/sangre , Adulto , Anciano , Estudios de Casos y Controles , Complemento C3b/análisis , Complemento C4b , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Puntaje de Gravedad del Traumatismo , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Heridas y Lesiones/complicaciones
7.
Am J Physiol Gastrointest Liver Physiol ; 304(3): G283-92, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23104558

RESUMEN

Ischemia-reperfusion (IR) injury causes a vigorous immune response that is amplified by complement activation, leading to local and remote tissue damage. Using MRL/lpr mice, which are known to experience accelerated tissue damage after mesenteric IR injury, we sought to evaluate whether complement inhibition mitigates organ damage. We found that complement depletion with cobra venom factor protected mice from local and remote lung tissue damage. Protection from injury was associated with less complement (C3) and membrane attack complex deposition, less neutrophil infiltration, and lower levels of local proinflammatory cytokine production. In addition, complement depletion was able to decrease the level of oxidative stress as measured by glutathione peroxidase 1 mRNA levels and superoxide dismutase activity. Furthermore, blockage of C5a receptor protected MRL/lpr mice from local tissue damage, but not from remote lung tissue damage. In conclusion, although treatments with cobra venom factor and C5a receptor antagonist were able to protect mice from local tissue damage, treatment with C5a receptor antagonist was not able to protect mice from remote lung tissue damage, implying that more factors contribute to the development of remote tissue damage after IR injury. These data also suggest that complement inhibition at earlier, rather than late, stages can have clinical benefit in conditions that are complicated with IR injury.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Lupus Eritematoso Sistémico/patología , Daño por Reperfusión/patología , Animales , Autoanticuerpos/farmacología , Complemento C5a/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/deficiencia , Venenos Elapídicos/farmacología , Femenino , Inmunohistoquímica , Mesenterio/patología , Ratones , Ratones Endogámicos MRL lpr , Infiltración Neutrófila/fisiología , Estrés Oxidativo/efectos de los fármacos , Adhesión en Parafina , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Circulación Esplácnica/fisiología
8.
J Surg Res ; 179(1): 153-67, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23122671

RESUMEN

BACKGROUND: Complement is invariably activated during trauma and contributes to tissue injury. Recombinant human decay-accelerating factor (DAF), a complement regulatory protein that inhibits both classical and alternative pathways, improves survival and reduces tissue damage in animal models of tissue injury. The extent to which DAF may facilitate resuscitation in hemorrhaged large animals is not known. METHODS: Male Yorkshire swine assigned to one of six groups were subjected to controlled, isobaric hemorrhage over 15 min to a target mean arterial pressure (MAP) of 35 mm Hg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of DAF or vehicle followed by Hextend resuscitation. Animals were observed for 3 h after hypotensive Hextend resuscitation. Survival, blood chemistry, and physiological parameters were recorded. Additionally, tissue from lung, small intestine, liver, and kidney were subjected to histopathologic evaluation and tissue deposition of complement proteins was determined by immunohistochemistry, dot-blot, and Western blot analyses. RESULTS: Administration of DAF (25 µg/kg) to animals subjected to hemorrhage prior to Hextend infusion significantly improved survival (73% versus 27%); protected gut, lung, liver, and kidney tissue from damage; and resulted in reduced resuscitation fluid requirements when compared with animals subjected to hemorrhage and resuscitation with Hextend alone. Animals treated with a higher dose of DAF (50 µg/kg) followed by Hextend fluid resuscitation did not experience the same benefit, suggesting a narrow therapeutic range for use of DAF as adjunct to Hextend fluid. CONCLUSION: DAF improved survival and reduced early Hextend fluid resuscitation requirements in swine subjected to hemorrhagic shock. These benefits are attributed to decreased complement deposition and limited organ damage.


Asunto(s)
Antígenos CD55/farmacología , Intestino Delgado/efectos de los fármacos , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Resucitación , Choque Hemorrágico/complicaciones , Animales , Relación Dosis-Respuesta a Droga , Fluidoterapia , Derivados de Hidroxietil Almidón/uso terapéutico , Intestino Delgado/patología , Riñón/efectos de los fármacos , Riñón/patología , Hígado/patología , Pulmón/patología , Masculino , Modelos Animales , Resucitación/métodos , Choque Hemorrágico/mortalidad , Tasa de Supervivencia , Porcinos , Resultado del Tratamiento
9.
Am J Physiol Gastrointest Liver Physiol ; 302(8): G888-97, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22301111

RESUMEN

Ischemia-reperfusion (I/R) injury is a leading cause of morbidity and mortality. A functional role for platelets in tissue damage after mesenteric I/R is largely unknown. The hypothesis that mesenteric I/R local and remote injury are platelet dependent was tested. Using a murine mesenteric I/R model, we demonstrate that platelets orchestrate remote lung tissue damage that follows mesenteric I/R injury and also contribute, albeit to a lesser degree, to local villi damage. While lung damage is delayed compared with villi damage, it increased over time and was characterized by accumulation of platelets in the pulmonary vasculature early, followed by alveolar capillaries and extravasation into the pulmonary space. Both villi and lung tissues displayed complement deposition. We demonstrate that villi and lung damage are reduced in mice made platelet deficient before I/R injury and that platelet transfusion into previously platelet-depleted mice before I/R increased both villi and lung tissue damage. Increased C3 deposition accompanied platelet sequestration in the lung, which was mostly absent in platelet-depleted mice. In contrast, C3 deposition was only minimally reduced on villi of platelet-depleted mice. Our findings position platelets alongside complement as a significant early upstream component that orchestrates remote lung tissue damage after mesenteric I/R and strongly suggest that reperfusion injury mitigating modalities should consider the contribution of platelets.


Asunto(s)
Plaquetas/fisiología , Daño por Reperfusión/patología , Circulación Esplácnica/fisiología , Lesión Pulmonar Aguda/patología , Animales , Complemento C3/fisiología , Citometría de Flujo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Intestinos/patología , Laparotomía , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Transfusión de Plaquetas , Circulación Pulmonar/fisiología
10.
Am J Physiol Gastrointest Liver Physiol ; 302(12): G1416-22, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22492694

RESUMEN

Tissue injury following ischemia-reperfusion (I/R) occurs as a consequence of actions of soluble factors and immune cells. Growing evidence supports a role for platelets in the manifestation of tissue damage following I/R. Spleen tyrosine kinase has been well documented to be important in lymphocyte activation and more recently in platelet activation. We performed experiments to evaluate whether inhibition of platelet activation through inhibition of spleen tyrosine kinase prevents tissue damage after mesenteric I/R injury. Platelets isolated from C57BL/6J mice fed with R788 for 10 days were transfused into C57BL/6J mice depleted of platelets 2 days before mesenteric I/R injury. Platelet-depleted mice transfused with platelets from R788-treated mice before mesenteric I/R displayed a significant reduction in the degree of remote lung damage, but with little change in the degree of local intestinal damage compared with control I/R mice. Transfusion of R788-treated platelets also decreased platelet sequestration, C3 deposition, and immunoglobulin deposition in lung, but not in the intestine, compared with control groups. These findings demonstrate that platelet activation is a requisite for sequestration in the pulmonary vasculature to mediate remote tissue injury after mesenteric I/R. The use of small-molecule inhibitors may be valuable to prevent tissue damage in remote organs following I/R injury.


Asunto(s)
Plaquetas/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/efectos de los fármacos , Oxazinas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Piridinas/farmacología , Daño por Reperfusión/metabolismo , Aminopiridinas , Animales , Plaquetas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/irrigación sanguínea , Intestinos/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Masculino , Ratones , Morfolinas , Recuento de Plaquetas , Transfusión de Plaquetas , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas , Quinasa Syk
11.
Am J Physiol Gastrointest Liver Physiol ; 301(6): G1020-30, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21903760

RESUMEN

Gut commensal bacteria play important roles in the development and homeostasis of intestinal immunity. However, the role of gut commensals in intestinal ischemia/reperfusion (I/R) injury is unclear. To determine the roles of gut commensal bacteria in intestinal IR injury, we depleted gut microbiota with a broad-spectrum antibiotic cocktail and performed mesenteric I/R (M I/R). First, we confirmed that antibiotic treatment completely depleted gut commensal bacteria and diminished the size of secondary lymphoid tissues such as the Peyer's patches. We next found that antibiotic treatment attenuated intestinal injury following M I/R. Depletion of gut commensal bacteria reduced the expression of Toll-like receptor (TLR)2 and TLR4 in the intestine. Both are well-known receptors for gram-positive and -negative bacteria. Decreased expression of TLR2 and TLR4 led to the reduction of inflammatory mediators, such as TNF, IL-6, and cyclooxygenase-2. Intestinal I/R injury is initiated when natural antibodies recognize neo-antigens that are revealed on ischemic cells and activate the complement pathway. Thus we evaluated complement and immunoglobulin (Ig) deposition in the damaged intestine and found that antibiotic treatment decreased the deposition of both C3 and IgM. Interestingly, we also found that the deposition of IgA also increased in the intestine following M I/R compared with control mice and that antibiotic treatment decreased the deposition of IgA in the damaged intestine. These results suggest that depletion of gut commensal bacteria decreases B cells, Igs, and TLR expression in the intestine, inhibits complement activation, and attenuates intestinal inflammation and injury following M I/R.


Asunto(s)
Enteritis/inmunología , Enteritis/microbiología , Intestinos/microbiología , Metagenoma/fisiología , Daño por Reperfusión/inmunología , Daño por Reperfusión/microbiología , Animales , Antibacterianos/farmacología , Ciego/inmunología , Ciego/microbiología , Ciego/patología , Proteínas del Sistema Complemento/metabolismo , Enteritis/patología , Homeostasis/inmunología , Inmunoglobulina A/metabolismo , Inmunoglobulina M/metabolismo , Intestinos/inmunología , Intestinos/patología , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Tejido Linfoide/patología , Masculino , Metagenoma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/patología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
12.
J Surg Res ; 167(2): e103-15, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20189588

RESUMEN

BACKGROUND: C-reactive protein (CRP) is an acute pro-inflammatory mediator that has been demonstrated to enhance ischemia/reperfusion (IR) injury by virtue of activating the complement system. CRP is able to interact with complement proteins such as C1q, complement factor H, and C4b-binding protein. Since complement activation is central in the expression of tissue injury following IR, we have investigated the effects of human decay-accelerating factor (DAF), a complement inhibitor, on CRP-potentiated complement activation and tissue injury in mice subjected to mesenteric IR. MATERIALS AND METHODS: Male C57B1/6 mice were allocated into eight groups: (1) Sham-operated group without IR injury; (2) CRP+Sham group; (3) IR group; (4) CRP+IR group; (5) DAF group; (6) CRP+DAF group; (7) IR+DAF group, and (8) CRP+IR+DAF group. Intestinal and lung injury, neutrophil infiltration, myeloperoxidase (MPO) expression, complement component deposition, and interleukin-6 (IL-6) production were assessed for each treatment group of mice. RESULTS: We report that administration of DAF significantly attenuates the CRP-enhanced intestinal injury as well as remote lung damages following acute mesenteric IR in mice, while DAF inhibits complement activation, suppresses neutrophil infiltration, and reduces IL-6 production. CONCLUSIONS: Our study suggests that inhibition complement activation with DAF may prove useful for the treatment of post-ischemic inflammatory injuries associated with an increased production of CRP.


Asunto(s)
Proteína C-Reactiva/metabolismo , Antígenos CD55/uso terapéutico , Mesenterio/metabolismo , Mesenterio/fisiopatología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Animales , Antígenos CD55/administración & dosificación , Antígenos CD55/farmacología , Complemento C5a/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Inyecciones Intravenosas , Interleucina-6/metabolismo , Masculino , Mesenterio/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infiltración Neutrófila/efectos de los fármacos , Peroxidasa/metabolismo , Daño por Reperfusión/fisiopatología
13.
J Trauma ; 71(1 Suppl): S151-60, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21795874

RESUMEN

BACKGROUND: Activation of complement system has been associated with tissue injury after hemorrhage and resuscitation in rats and swine. This study investigated whether administration of human recombinant decay-accelerating factor (DAF; a complement regulatory protein that inhibits classical and alternative pathways) reduces tissue damage in a porcine model of hemorrhagic shock. METHODS: Male Yorkshire swine assigned to four groups were subjected to controlled, isobaric hemorrhage over 15 minutes to a target mean arterial pressure of 35 mm Hg. Hypotension was maintained for 20 minutes followed by a bolus intravenous injection of DAF or vehicle and then animals were observed for 200 minutes. Blood chemistry and physiologic parameters were recorded. Tissue samples from lung and small intestine were subjected to histopathological evaluation and detection of tissue deposition of complement proteins by immunohistochemistry and Western blot analyses. RESULTS: Administration of DAF significantly reduced intestinal and lung tissue damage in a dose-dependent manner (5, 25, and 50 µg/kg). In addition, DAF treatment improved hemorrhage-induced hyperkalemia. The protective effects of DAF appear to be related to its ability to reduce tissue complement activation and deposition on affected tissues. CONCLUSIONS: DAF treatment decreased tissue complement activation and deposition in hemorrhaged animals and attenuated tissue damage at 200 minutes after treatment. The observed beneficial effects of DAF treatment on tissue injury after 20 minutes of severe hypotension presents an attractive model of small volume resuscitation, particularly in situations with a restrictive medical logistical footprint such as far-forward access to first responders in the battlefield or in remote rural or mountainous environments.


Asunto(s)
Antígenos CD55/uso terapéutico , Hiperpotasemia/prevención & control , Intestinos/patología , Pulmón/patología , Choque Hemorrágico/tratamiento farmacológico , Animales , Western Blotting , Activación de Complemento/efectos de los fármacos , Hemodinámica , Humanos , Hiperpotasemia/etiología , Intestinos/efectos de los fármacos , Pulmón/efectos de los fármacos , Masculino , Proteínas Recombinantes/uso terapéutico , Resucitación/métodos , Choque Hemorrágico/complicaciones , Choque Hemorrágico/patología , Porcinos , Factores de Tiempo
14.
Am J Physiol Gastrointest Liver Physiol ; 299(2): G391-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20522642

RESUMEN

Reperfusion injury to tissue following an ischemic event occurs as a consequence of an acute inflammatory response that can cause significant morbidity and mortality. Components of both the innate (complement, immunoglobulin, monocytes, and neutrophils) and adaptive (B and T lymphocytes) immune systems have been demonstrated to mediate tissue injury. Spleen tyrosine kinase (Syk) is responsible for membrane-mediated signaling in various cell types including B lymphocytes, macrophages, and T cells. We investigated the ability of a small drug Syk inhibitor, R788, to protect mice against mesenteric ischemia-reperfusion (I/R)-induced local (intestine) and remote lung injury. Mice were fed with chow containing a Syk inhibitor for 6 days before the performance of intestinal I/R, which resulted in silencing of the expression of the active phosphorylated Syk. Syk inhibition significantly suppressed both local and remote lung injury. The beneficial effect was associated with reduced IgM and complement 3 deposition in the tissues and significant reduction of polymorphonuclear cell infiltration. Our data place Syk upstream of events leading to the binding of natural antibodies to the ischemia-conditioned tissues and urge the consideration of the use of Syk inhibitors in the prevention or improvement of tissue injury of organs exposed to ischemia or hypoperfusion.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Circulación Esplácnica , Aminopiridinas , Animales , Complemento C3/antagonistas & inhibidores , Complemento C3/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Intestinos/irrigación sanguínea , Intestinos/efectos de los fármacos , Intestinos/patología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Morfolinas , Infiltración Neutrófila , Oxazinas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Circulación Pulmonar , Piridinas/farmacología , Pirimidinas , Receptores de Superficie Celular/metabolismo , Daño por Reperfusión/metabolismo , Quinasa Syk
15.
J Neuroinflammation ; 7: 24, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20380727

RESUMEN

BACKGROUND: Activated complement system is known to mediate neuroinflammation and neurodegeneration following exposure to hypoxic-ischemic insults. Therefore, inhibition of the complement activation cascade may represent a potential therapeutic strategy for the management of ischemic brain injury. Decay-accelerating factor (DAF, also known as CD55) inhibits complement activation by suppressing the function of C3/C5 convertases, thereby limiting local generation or deposition of C3a/C5a and membrane attack complex (MAC or C5b-9) production. The present study investigates the ability of DAF to protect primary cultured neuronal cells subjected to sodium cyanide (NaCN)-induced hypoxia from degeneration and apoptosis. METHODS: Cultured primary cortical neurons from embryonic Sprague-Dawley rats were assigned one of four groups: control, DAF treatment alone, hypoxic, or hypoxic treated with DAF. Hypoxic cultures were exposed to NaCN for 1 hour, rinsed, followed by 24 hour exposure to 200 ng/ml of recombinant human DAF in normal medium. Human DAF was used in the present study and it has been shown to effectively regulate complement activation in rats. Neuronal cell function, morphology and viability were investigated by measuring plateau depolarization potential, counting the number dendritic spines, and observing TUNEL and MTT assays. Complement C3, C3a, C3a receptor (R) production, C3a-C3aR interaction and MAC formation were assessed along with the generation of activated caspase-9, activated caspase-3, and activated Src. RESULTS: When compared to controls, hypoxic cells had fewer dendritic spines, reduced plateau depolarization accompanied by increased apoptotic activity and accumulation of MAC, as well as up-regulation of C3, C3a and C3aR, enhancement of C3a-C3aR engagement, and elevated caspase and Src activity. Treatment of hypoxic cells with 200 ng/ml of recombinant human DAF resulted in attenuation of neuronal apoptosis and exerted significant protection against neuronal dendritic spine loss and plateau depolarization reduction. Furthermore, treatment with DAF resulted in decreased accumulation of C3a, MAC, C3a-C3aR interaction, caspase-9, activated caspase-3, and pTyr416-Src (activated Src) tyrosine kinase. CONCLUSION: DAF was found to reduce neuronal cell death and apoptosis in NaCN induced hypoxia. This effect is attributed to the ability of DAF to limit complement activation and inhibit the activity of Src and caspases 9 and 3. This study supports the inhibiting of complement as a neuroprotective strategy against CNS ischemia/reperfusion injury.


Asunto(s)
Antígenos CD55/farmacología , Hipoxia de la Célula/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Complemento C3a/metabolismo , Proteínas del Sistema Complemento/fisiología , Dendritas/fisiología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de Complemento/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
16.
Shock ; 53(1): 16-23, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30998651

RESUMEN

BACKGROUND: Activation of the complement system and complement deposition on red blood cells (RBCs) contribute to organ damage in trauma. We conducted a prospective study in subjects with traumatic injuries to determine the pattern of complement deposition on RBC and whether they are associated with clinical outcomes. METHOD: A total of 124 trauma patients and 42 healthy controls were enrolled in this prospective study. RBC and sera were collected at 0, 6, 24, and 72 h from trauma patients and healthy controls during a single draw. Presence of C4d, C3d, C5b-9, phosphorylation of band 3 and production of nitric oxide were analyzed by flow cytometry. RESULTS: RBC from trauma patients at all time points up to 24 h displayed significantly higher deposition of C4d on their RBC membrane as compared with healthy donors. Incubation of normal RBC with sera from trauma patients resulted in significant increase of C4d deposition (at 0, 6, 24, and 72 h), C5b-9 deposition (at 0 and 6 h), phosphorylation of band 3 (at 0 and 24 h), and nitric oxide production up to 24 h compared with sera from healthy subjects. Deposition of C4d and C5b-9 in patients with an Injury Severity Score (ISS) of 9 and above remained elevated up to 72 h. CONCLUSIONS: Our study demonstrates that the presence of C4d, C3d, and C5b-9 on the surface of RBC is linked to increased phosphorylation of band 3 and increased production of nitric oxide. Deposition of C4d and C5b-9 decreased faster over course of 3-day study in subjects with ISS less than 9.


Asunto(s)
Biomarcadores/metabolismo , Proteínas del Sistema Complemento/metabolismo , Eritrocitos/metabolismo , Heridas y Lesiones/metabolismo , Adulto , Anciano , Activación de Complemento/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Estudios Prospectivos
17.
Clin Immunol ; 130(3): 313-21, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19058762

RESUMEN

Elements of the innate and adaptive immune response have been implicated in the development of tissue damage after ischemic reperfusion (I/R). Here we demonstrate that T cells infiltrate the intestine of C57BL/6 mice subjected to intestinal I/R during the first hour of reperfusion. The intensity of the T cell infiltration was higher in B6.MRL/lpr mice subjected to intestinal I/R and reflected more severe tissue damage than that observed in control mice. Depletion of T cells limited I/R damage in B6.MRL/lpr mice, whereas repletion of B6.MRL/lpr lymph node-derived T cells into the I/R-resistant Rag-1(-/-) mouse reconstituted tissue injury. The tissue-infiltrating T cells were found to produce IL-17. Finally, IL-23 deficient mice, which are known not to produce IL-17, displayed significantly less intestinal damage when subjected to I/R. Our data assign T cells a major role in intestinal I/R damage by virtue of producing the pro-inflammatory cytokine IL-17.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Linfocitos T CD4-Positivos/inmunología , Interleucina-17/inmunología , Intestinos , Daño por Reperfusión/fisiopatología , Animales , Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Intestinos/inmunología , Intestinos/lesiones , Intestinos/fisiopatología , Isquemia/inmunología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Noqueados , Daño por Reperfusión/inmunología
18.
Acute Med Surg ; 6(4): 329-335, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31592318

RESUMEN

Trauma remains a major cause of death throughout the world, especially for patients younger than 45 years. Due to rapid advances in clinical management, both in the acute and prehospital settings, trauma patients survive devastating injuries at unprecedented rates. However, these patients can often face life threatening complications that stem from the robust innate immune response induced by severe hemorrhage, leading to further tissue injury rather than repair. The complement and coagulation cascades are key mediators in this disordered reaction, which includes the development of trauma-induced coagulopathy. There is increasing evidence that cross-talk between these two pathways allows rapid amplification of their otherwise targeted responses and contributes to overwhelming and prolonged systemic inflammation. In this article, we summarize the initial steps of innate immune response to trauma and review the complex complement and coagulation cascades, as well as how they interact with each other. Despite progress in understanding these cascades, effective therapeutic targets have yet to be found and further research is needed both to improve survival rates as well as decrease associated morbidity.

19.
Front Immunol ; 8: 1046, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28928734

RESUMEN

C3a is important in the regulation of the immune response as well as in the development of organ inflammation and injury. Furthermore, C3a contributes to liver regeneration but its role in intestinal stem cell function has not been studied. We hypothesized that C3a is important for intestinal repair and regeneration. Intestinal organoid formation, a measure of stem cell capacity, was significantly limited in C3-deficient and C3a receptor (C3aR) 1-deficient mice while C3a promoted the growth of organoids from normal mice by supporting Wnt-signaling but not from C3aR1-deficient mice. Similarly, the presence of C3a in media enhanced the expression of the intestinal stem cell marker leucine-rich repeat G-protein-coupled receptor 5 (Lgr5) and of the cell proliferation marker Ki67 in organoids formed from C3-deficient but not from C3aR1-deficient mice. Using Lgr5.egfp mice we showed significant expression of C3 in Lgr5+ intestinal stem cells whereas C3aR1 was expressed on the surface of various intestinal cells. C3 and C3aR1 expression was induced in intestinal crypts in response to ischemia/reperfusion injury. Finally, C3aR1-deficient mice displayed ischemia/reperfusion injury comparable to control mice. These data suggest that C3a through interaction with C3aR1 enhances stem cell expansion and organoid formation and as such may have a role in intestinal regeneration.

20.
Shock ; 46(3 Suppl 1): 177-88, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27405065

RESUMEN

Complement system activation is recognized as a deleterious component of the mammalian physiological response to traumatic injury with severe hemorrhage (TH). Female Yorkshire swine were subjected to a simulated austere prehospital battlefield scenario. Each animal underwent controlled hemorrhage of 22 mL/kg at 100 mL/min rate for approximately 10 min followed by soft tissue injury, femur fracture, and spleen injury. Subsequent blood loss was uncontrolled. Twenty-eight minutes postinjury the animals were randomized into treatment or no treatment with recombinant human C1 esterase inhibitor (C1INH) (500 IU/kg, n = 11) and into receiving or not permissive hypotensive resuscitation (n = 14) with infusion of 45 mL/kg lactated Ringer's solution (2× blood lost). Observation and animal maintenance continued for 6 h at which time the animals had either expired or were euthanized. Heart, lung, and small intestine tissue samples were collected. Pharmacokinetic, hemodynamic, and metabolic parameters as well as survival time, plasma complement activity and tissue deposition, cytokine levels, and tissue injury were determined. We found that administration of C1INH protected tissues from damage, reduced the levels of inflammatory cytokines, and improved blood chemistry. Immunohistochemical analyses revealed that C1INH administration following TH markedly reduced complement activation and deposition in tissues. Importantly, C1INH administration prolonged survival of animals particularly in those which received resuscitation fluid infusion. Our data urge early administration of C1INH to limit organ damage and prolong survival of those injured in the battlefield.


Asunto(s)
Proteína Inhibidora del Complemento C1/uso terapéutico , Resucitación/métodos , Guerra , Animales , Activación de Complemento/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fracturas del Cuello Femoral/tratamiento farmacológico , Fracturas del Cuello Femoral/terapia , Fluidoterapia , Hemodinámica/efectos de los fármacos , Hemorragia/tratamiento farmacológico , Hemorragia/terapia , Soluciones Isotónicas/uso terapéutico , Lactato de Ringer , Choque Hemorrágico/tratamiento farmacológico , Choque Hemorrágico/terapia , Traumatismos de los Tejidos Blandos/tratamiento farmacológico , Traumatismos de los Tejidos Blandos/terapia , Bazo/efectos de los fármacos , Bazo/lesiones , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA