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










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(5)2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38473750

RESUMO

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.


Assuntos
Acidose , Transtornos da Coagulação Sanguínea , Choque Hemorrágico , Humanos , Feminino , Suínos , Animais , Insuficiência de Múltiplos Órgãos , Hemorragia , Citocinas
2.
Shock ; 53(1): 16-23, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30998651

RESUMO

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.


Assuntos
Biomarcadores/metabolismo , Proteínas do Sistema Complemento/metabolismo , Eritrócitos/metabolismo , Ferimentos e Lesões/metabolismo , Adulto , Idoso , Ativação do Complemento/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Estudos Prospectivos
3.
Acute Med Surg ; 6(4): 329-335, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31592318

RESUMO

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.

4.
J Immunol ; 199(8): 2921-2929, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28877988

RESUMO

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.


Assuntos
Interleucina-17/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/patologia , Linfócitos/imunologia , Traumatismo por Reperfusão/imunologia , Animais , Anticorpos Bloqueadores/administração & dosagem , Autoanticorpos/metabolismo , Células Cultivadas , Ativação do Complemento , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Humanos , Imunidade Inata , Interleucina-17/genética , Artéria Mesentérica Superior/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Receptores de Interleucina/genética
5.
Front Immunol ; 8: 1046, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28928734

RESUMO

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.

6.
J Immunol ; 198(2): 788-797, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27913632

RESUMO

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.


Assuntos
Complemento C3/metabolismo , Isquemia Mesentérica/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Western Blotting , Células CACO-2 , Catepsinas/metabolismo , Complemento C3/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Isquemia Mesentérica/imunologia , Isquemia Mesentérica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia
7.
Shock ; 46(3 Suppl 1): 177-88, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27405065

RESUMO

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.


Assuntos
Proteína Inibidora do Complemento C1/uso terapêutico , Ressuscitação/métodos , Guerra , Animais , Ativação do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fraturas do Colo Femoral/tratamento farmacológico , Fraturas do Colo Femoral/terapia , Hidratação , Hemodinâmica/efeitos dos fármacos , Hemorragia/tratamento farmacológico , Hemorragia/terapia , Soluções Isotônicas/uso terapêutico , Lactato de Ringer , Choque Hemorrágico/tratamento farmacológico , Choque Hemorrágico/terapia , Lesões dos Tecidos Moles/tratamento farmacológico , Lesões dos Tecidos Moles/terapia , Baço/efeitos dos fármacos , Baço/lesões , Suínos
8.
Mil Med ; 179(8 Suppl): 34-40, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25102547

RESUMO

BACKGROUND: Blast-induced ocular trauma is a frequent cause of morbidity for survivors of improvised explosive devices. Blast overpressure (BOP) of 120 ± 7 KPa has been shown to cause damage to lungs, brain, and gut in a rat model; however, the effects of BOP on ocular tissues have not been characterized. To elucidate the pathophysiology of blast-induced ocular trauma, ocular tissues from rats subjected to blast were examined for evidence of apoptosis by the detection of activated caspase 3 and TUNEL assay in their ocular tissues. METHODS: A compressed air shock tube was used to deliver 120 ± 7 KPa of BOP for duration of 2 msec to the right side of the rats. Rats were then euthanized at specific time points after blast exposure (3 hours, 24 hours, 48 hours). Ocular tissues were processed for immunohistochemistry to detect activated caspase 3 and TUNEL assay. Tissues were evaluated for relative levels of positive signal as compared to nonblast exposed controls. RESULTS: Activated caspase 3 was detected in the optic nerve, ganglion layer, and inner nuclear layer post blast exposure. At 24 and 48 hours, the inner nuclear layer from the right side had more cells with activated caspase 3. In the optic nerve, the highest levels of activated caspase 3 were detected on the right side at 24 hours post blast. CONCLUSION: BOP of 120 ± 7 KPa induces optic neuropathy and retinal damage. In both the optic nerve and retina, caspase 3 was activated in the right and left sides following blast exposure. The results of this study reveal that blast exposure induces apoptosis in both the optic nerve and retinal tissues.


Assuntos
Traumatismos por Explosões/fisiopatologia , Traumatismos Oculares/fisiopatologia , Traumatismos do Nervo Óptico/fisiopatologia , Retina/lesões , Animais , Apoptose , Caspase 3/análise , Masculino , Traumatismos do Nervo Óptico/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/química
9.
Crit Care Med ; 42(5): e364-72, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24448198

RESUMO

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.


Assuntos
Cálcio/sangue , Ativação do Complemento/fisiologia , Eritrócitos/metabolismo , Óxido Nítrico/sangue , Fragmentos de Peptídeos/sangue , Ferimentos e Lesões/sangue , Adulto , Idoso , Estudos de Casos e Controles , Complemento C3b/análise , Complemento C4b , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Escala de Gravidade do Ferimento , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Ferimentos e Lesões/complicações
10.
Glycobiology ; 24(4): 379-91, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24440830

RESUMO

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.


Assuntos
Lectinas Tipo C/metabolismo , Leucócitos Mononucleares/metabolismo , Receptores de Complemento/metabolismo , Explosão Respiratória , Transdução de Sinais , Humanos
11.
Acta Neuropathol Commun ; 1: 52, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-24252631

RESUMO

BACKGROUND: Blast-induced neurotrauma (BINT) is the signature life threatening injury of current military casualties. Neuroinflammation is a key pathological occurrence of secondary injury contributing to brain damage after blast injury. We have recently demonstrated that blast-triggered complement activation and cytokine release are associated with BINT. Here, we evaluated if administration of the complement inhibitor recombinant human decay-accelerating factor (rhDAF) is beneficial on neuroinflammation and neurodegeneration in a rat model of moderate BINT. Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI. RESULT: Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. CONCLUSION: These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI.


Assuntos
Traumatismos por Explosões/complicações , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/etiologia , Antígenos CD55/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Neuroimunomodulação/efeitos dos fármacos , Pressão , Distribuição Aleatória , Ratos Sprague-Dawley , Fatores de Tempo , Resultado do Tratamento
12.
Proc Natl Acad Sci U S A ; 110(35): 14348-53, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23942120

RESUMO

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.


Assuntos
Endotélio Vascular/metabolismo , Isquemia Mesentérica/metabolismo , Trombospondinas/metabolismo , Animais , Endotélio Vascular/patologia , Junções Intercelulares/efeitos dos fármacos , Isquemia Mesentérica/tratamento farmacológico , Camundongos , Ligação Proteica , Traumatismo por Reperfusão/prevenção & controle , Trombospondinas/farmacologia
13.
J Trauma Acute Care Surg ; 75(2 Suppl 2): S115-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23883895

RESUMO

BACKGROUND: The implementation of a human research protection program in Afghanistan and the mobilization of the combat casualty research team have made it possible to design and efficiently conduct multifaceted, multisite, and prospective research studies in a combat environment. Still, to conduct research in such an environment, several unique challenges must be overcome. METHODS: This article describes the development and conduct of three ongoing trauma-related biomedical research studies in Afghanistan, highlighting the challenges and lessons learned within the context of these studies. RESULTS: Key challenges include the process of developing and getting approval for in-theater research protocols, the informed consent process, and logistics of conducting a biomedical research study in an austere environment. Despite these challenges, important lessons learned that can contribute to the success of a protocol include the need for clear operating procedures, judicious selection for which data points must be collected in-theater, and the importance anticipating the "fog and friction" of war. CONCLUSION: As we continue the journey toward more sophisticated research capabilities in combat, this article will help inform the design and conduct of future research performed in a theater of war. Conducting biomedical research in a combat zone is an important but difficult element of military medicine.


Assuntos
Campanha Afegã de 2001- , Pesquisa Biomédica/métodos , Medicina Militar/métodos , Pesquisa Biomédica/organização & administração , Humanos , Consentimento Livre e Esclarecido , Medicina Militar/organização & administração , Estados Unidos , Ferimentos e Lesões/cirurgia , Ferimentos e Lesões/terapia
14.
Am J Physiol Gastrointest Liver Physiol ; 304(3): G283-92, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23104558

RESUMO

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.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Lúpus Eritematoso Sistêmico/patologia , Traumatismo por Reperfusão/patologia , Animais , Autoanticorpos/farmacologia , Complemento C5a/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Proteínas do Sistema Complemento/deficiência , Venenos Elapídicos/farmacologia , Feminino , Imuno-Histoquímica , Mesentério/patologia , Camundongos , Camundongos Endogâmicos MRL lpr , Infiltração de Neutrófilos/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Inclusão em Parafina , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Circulação Esplâncnica/fisiologia
15.
J Surg Res ; 179(1): 153-67, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23122671

RESUMO

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.


Assuntos
Antígenos CD55/farmacologia , Intestino Delgado/efeitos dos fármacos , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Ressuscitação , Choque Hemorrágico/complicações , Animais , Relação Dose-Resposta a Droga , Hidratação , Derivados de Hidroxietil Amido/uso terapêutico , Intestino Delgado/patologia , Rim/efeitos dos fármacos , Rim/patologia , Fígado/patologia , Pulmão/patologia , Masculino , Modelos Animais , Ressuscitação/métodos , Choque Hemorrágico/mortalidade , Taxa de Sobrevida , Suínos , Resultado do Tratamento
16.
PLoS One ; 7(7): e39934, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792197

RESUMO

The robust inflammatory response that occurs during ischemia reperfusion (IR) injury recruits factors from both the innate and adaptive immune systems. However the contribution of platelets and their products such as Platelet Factor 4 (PF4; CXCL4), during the pathogenesis of IR injury has not been thoroughly investigated. We show that a deficiency in PF4 protects mice from local and remote tissue damage after 30 minutes of mesenteric ischemia and 3 hours of reperfusion in PF4-/- mice compared to control B6 mice. This protection was independent from Ig or complement deposition in the tissues. However, neutrophil and monocyte infiltration were decreased in the lungs of PF4-/- mice compared with B6 control mice. Platelet-depleted B6 mice transfused with platelets from PF4-/- mice displayed reduced tissue damage compared with controls. In contrast, transfusion of B6 platelets into platelet depleted PF4-/- mice reconstituted damage in both intestine and lung tissues. We also show that PF4 may modulate the release of IgA. Interestingly, we show that PF4 expression on intestinal epithelial cells is increased after IR at both the mRNA and protein levels. In conclusion, these findings demonstrate that may PF4 represent an important mediator of local and remote tissue damage.


Assuntos
Artérias Mesentéricas/patologia , Mesentério/metabolismo , Mesentério/patologia , Fator Plaquetário 4/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Imunoglobulinas/imunologia , Imunoglobulinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Knockout , Monócitos/patologia , Infiltração de Neutrófilos , Fator Plaquetário 4/genética , Transfusão de Plaquetas , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/terapia
17.
Shock ; 38(1): 82-91, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22683724

RESUMO

Activation of the complement system has been associated with tissue injury after hemorrhage and resuscitation in animals. We investigated whether administration of recombinant human C1-esterase inhibitor (rhC1-INH), a regulator of complement and contact activation systems, reduces tissue damage and cytokine release and improves metabolic acidosis in a porcine model of hemorrhagic shock. Male Yorkshire swine were assigned to experimental groups and subjected to controlled, isobaric hemorrhage to a target mean arterial pressure of 35 mmHg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of rhC1-INH or vehicle; animals were then observed for 3 h. Blood chemistry and physiologic parameters were recorded. Lung and small intestine tissue samples were subjected to histopathologic evaluation and immunohistochemistry to determine the extent of injury and deposition of complement proteins. Cytokine levels and quantitative assessment of renal and hepatic function were measured via enzyme-linked immunosorbent assay and chemistry analyzer, respectively. Pharmacokinetics of rhC1-INH revealed dose proportionality for maximum concentration, half-life, and the time span in which the functional C1-INH level was greater than 1 IU/mL. Recombinant human C1-INH significantly reduced renal, intestinal, and lung tissue damage in a dose-dependent manner (100 and 250 IU/kg). In addition, rhC1-INH (250 IU/kg) markedly improved hemorrhage-induced metabolic acidosis and circulating tumor necrosis factor α. The tissue-protective effects of rhC1-INH appear to be related to its ability to reduce tissue complement activation and deposition. Recombinant human C1-INH decreased tissue complement activation and deposition in hemorrhaged animals, improved metabolic acidosis, reduced circulating tumor necrosis factor α, and attenuated tissue damage in this model. The observed beneficial effects of rhC1-INH treatment on tissue injury 20 min into severe hypotension present an attractive model of low-volume resuscitation, particularly in situations with a restrictive medical logistical footprint.


Assuntos
Proteína Inibidora do Complemento C1/uso terapêutico , Inativadores do Complemento/uso terapêutico , Choque Hemorrágico/tratamento farmacológico , Acidose/tratamento farmacológico , Acidose/etiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Proteína Inibidora do Complemento C1/administração & dosagem , Proteína Inibidora do Complemento C1/farmacologia , Inativadores do Complemento/administração & dosagem , Inativadores do Complemento/farmacologia , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Infusões Intravenosas , Enteropatias/etiologia , Enteropatias/prevenção & controle , Intestino Delgado/metabolismo , Rim/efeitos dos fármacos , Rim/fisiopatologia , Pulmão/metabolismo , Pneumopatias/etiologia , Pneumopatias/prevenção & controle , Masculino , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/prevenção & controle , Choque Hemorrágico/sangue , Choque Hemorrágico/complicações , Choque Hemorrágico/fisiopatologia , Sus scrofa , Fator de Necrose Tumoral alfa/metabolismo
18.
Am J Physiol Gastrointest Liver Physiol ; 302(12): G1416-22, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22492694

RESUMO

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.


Assuntos
Plaquetas/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pulmão/efeitos dos fármacos , Oxazinas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Piridinas/farmacologia , Traumatismo por Reperfusão/metabolismo , Aminopiridinas , Animais , Plaquetas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/irrigação sanguínea , Intestinos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Masculino , Camundongos , Morfolinas , Contagem de Plaquetas , Transfusão de Plaquetas , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas , Quinase Syk
19.
J Neurol Sci ; 318(1-2): 146-54, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22537900

RESUMO

Blast-induced neurotrauma (BINT) is a major medical concern yet its etiology is largely undefined. Complement activation may play a role in the development of secondary injury following traumatic brain injury; however, its role in BINT is still undefined. The present study was designed to characterize the complement system and adaptive immune-inflammatory responses in a rat model of moderate BINT. Anesthetized rats were exposed to a moderate blast (120 kPa) using an air-driven shock tube. Brain tissue injury, systemic and local complement, cerebral edema, inflammatory cell infiltration, and pro-inflammatory cytokine production were measured at 0.5, 3, 48, 72, 120, and 168 h. Injury to brain tissue was evaluated by histological evaluation. Systemic complement was measured via ELSIA. The remaining measurements were determined by immunohistoflourescent staining. Moderate blast triggers moderate brain injuries, elevated levels of local brain C3/C5b-9 and systemic C5b-9, increased leukocyte infiltration, unregulated tumor necrosis factor alpha (TNFα), and aquaporin-4 in rat brain cortex at 3- and 48-hour post blast. Early immune-inflammatory response to BINT involves complement and TNFα, which correlates with hippocampus and cerebral cortex damage. Complement and TNFα activation may be a novel therapeutic target for reducing the damaging effects of BINT inflammation.


Assuntos
Traumatismos por Explosões/fisiopatologia , Lesões Encefálicas/fisiopatologia , Ativação do Complemento/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Traumatismos por Explosões/imunologia , Traumatismos por Explosões/patologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
PLoS One ; 7(2): e32260, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384195

RESUMO

Several innate and adaptive immune cell types participate in ischemia/reperfusion induced tissue injury. Amongst them, platelets have received little attention as contributors in the process of tissue damage after ischemia reperfusion (I/R) injury. It is currently unknown whether platelets participate through the immunologically important molecules including, CD40 and when activated, CD154 (CD40L), in the pathogenesis of I/R injury. We hypothesized that constitutive expression of CD40 and activation-induced expression of CD154 on platelets mediate local mesenteric and remote lung tissue damage after I/R injury. Wild type (WT; C57BL/6J), CD40 and CD154 deficient mice underwent mesenteric ischemia for 30 minutes followed by reperfusion for 3 hours. WT mice subjected to mesenteric I/R injury displayed both local intestinal and remote lung damage. In contrast, there was significantly less intestinal damage and no remote lung injury in CD40 and CD154 deficient mice when compared to WT mice. Platelet-depleted WT mice transfused with platelets from CD40 or CD154 deficient mice failed to reconstitute remote lung damage. In contrast, when CD40 or CD154 deficient mice were transfused with WT platelets lung tissue damage was re-established. Together, these findings suggest that multiple mechanisms are involved in local and remote tissue injury and also identify platelet-expressed CD40 and/or CD154 as mediators of remote tissue damage.


Assuntos
Antígenos CD40/fisiologia , Ligante de CD40/fisiologia , Traumatismo por Reperfusão/metabolismo , Animais , Plaquetas/citologia , Plaquetas/metabolismo , Antígenos CD40/genética , Ligante de CD40/genética , Proteínas do Sistema Complemento , Endotoxinas/metabolismo , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica/métodos , Intestinos/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...