Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 329
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209943

ABSTRACT

Severe or major burns induce a pathophysiological, immune, and inflammatory response that can persist for a long time and affect morbidity and mortality. Severe burns are followed by a "hypermetabolic response", an inflammatory process that can be extensive and become uncontrolled, leading to a generalized catabolic state and delayed healing. Catabolism causes the upregulation of inflammatory cells and innate immune markers in various organs, which may lead to multiorgan failure and death. Burns activate immune cells and cytokine production regulated by damage-associated molecular patterns (DAMPs). Trauma has similar injury-related immune responses, whereby DAMPs are massively released in musculoskeletal injuries and elicit widespread systemic inflammation. Hemorrhagic shock is the main cause of death in trauma. It is hypovolemic, and the consequence of volume loss and the speed of blood loss manifest immediately after injury. In burns, the shock becomes evident within the first 24 h and is hypovolemic-distributive due to the severely compromised regulation of tissue perfusion and oxygen delivery caused by capillary leakage, whereby fluids shift from the intravascular to the interstitial space. In this review, we compare the pathophysiological responses to burns and trauma including their associated clinical patterns.


Subject(s)
Alarmins/metabolism , Burns/immunology , Shock, Hemorrhagic/immunology , Cytokines/metabolism , Gene Expression Regulation , Humans , Mitochondria/metabolism
2.
J Surg Res ; 247: 453-460, 2020 03.
Article in English | MEDLINE | ID: mdl-31668606

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a frequent complication after severe trauma. Lung-protective ventilation strategies and damage control resuscitation have been proposed for the prevention of ALI; however, there are no clinical or laboratory parameters to predict who is at risk of developing ALI after trauma. In the present study, we explored pulmonary inflammatory markers as a potential predictor of ALI using a porcine model of hemorrhagic shock. MATERIALS AND METHODS: Female swine were randomized to mechanical ventilation with low tidal volume (VT) (6 mL/kg) or high VT (12 mL/kg). After equilibration, animals underwent pressure-controlled hemorrhage (mean arterial pressure [MAP] 35 ± 5 mmHg) for 1 h, followed by resuscitation with fresh whole blood or Hextend. They were maintained at MAP of 50 ± 5 mmHg for 3 h in the postresuscitation phase. Bronchoalveolar lavage fluids were collected hourly and analyzed for inflammatory markers. Lung samples were taken, and porcine neutrophil antibody staining was used to evaluate the presence of neutrophils. ELISA evaluated serum porcine surfactant protein D levels. Sham animals were used as negative controls. RESULTS: Pigs that underwent hemorrhagic shock had higher heart rates, lower cardiac output, lower MAPs, and worse acidosis compared with sham at the early time points (P < 0.05 each). There were no significant differences in central venous pressure or pulmonary capillary wedge pressure between groups. Pulmonary neutrophil infiltration, as defined by neutrophil antibody staining on lung samples, was greater in the shock groups regardless of resuscitation fluid (P < 0.05 each). Bronchoalveolar lavage fluid neutrophil levels were not different between groups. There were no differences in levels of porcine surfactant protein D between groups at any time points, and the levels did not change over time in each respective group. CONCLUSIONS: Our study demonstrates the reproducibility of a porcine model of hemorrhagic shock that is consistent with physiologic changes in humans in hemorrhagic shock. Pulmonary neutrophil infiltration may serve as an early marker for ALI; however, the practicality of this finding has yet to be determined.


Subject(s)
Acute Lung Injury/diagnosis , Neutrophils/immunology , Shock, Hemorrhagic/complications , Acute Lung Injury/immunology , Acute Lung Injury/physiopathology , Acute Lung Injury/prevention & control , Animals , Blood Transfusion , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cardiac Output/immunology , Disease Models, Animal , Female , Heart Rate/immunology , Humans , Lung/cytology , Lung/immunology , Lung/pathology , Neutrophil Infiltration , Predictive Value of Tests , Prognosis , Pulmonary Surfactant-Associated Protein D/analysis , Pulmonary Surfactant-Associated Protein D/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Reproducibility of Results , Respiration, Artificial/instrumentation , Respiration, Artificial/methods , Resuscitation/methods , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/therapy , Sus scrofa , Time Factors
3.
J Surg Res ; 256: 220-230, 2020 12.
Article in English | MEDLINE | ID: mdl-32711179

ABSTRACT

BACKGROUND: Immune dysfunction is associated with posthemorrhagic shock mesenteric lymph (PHSML) return. To determine the proliferation and cytokine production capacity of CD4+ T lymphocytes, the effect of PHSML drainage on spleen CD4+ T lymphocytes in a mouse model of hemorrhagic shock was assessed. METHODS: The normal spleen CD4+ T lymphocytes were in vitro incubated with either drained normal mesenteric lymph (NML), PHSML during hypotension (PHSML-H), or PHSML from 0 h to 3 h after resuscitation (PHSML-R) to verify direct proliferation effects of PHSML. RESULTS: Hemorrhagic shock led to reduction of proliferation and mRNA expression of interleukin 2 (IL-2) and IL-2 receptor in CD4+ T lymphocytes and to decrease in IL-2 and interferon γ (IFN-γ) levels in supernatants. In contrast, the interleukin-4 levels were increased. These effects were reversed by PHSML drainage. Moreover, NML incubation promoted CD4+ T lymphocyte proliferation, whereas both PHSML-H and PHSML-R treatment had a biphasic effects on CD4+ T lymphocyte proliferation, exhibiting an enhanced effect at early stages and an inhibitory effect at later stages. Compared with NML, PHSML-H increased IL-2 expression at 12 h, but decreased expression of both IL-2 and IFN-γ at 24 h. By contrast, PHSML-R induced significant increases in IL-2 and IFN-γ levels at 24 h. Interleukin-4 expression in CD4+ T lymphocytes was reduced at 12 h, but augmented at 24 h after incubation with either PHSML-H or PHSML-R. CONCLUSIONS: The results indicate that PHSML has a direct inhibitory effect on CD4+ T lymphocyte proliferation that induces an inflammatory response, which is associated with cellular immune dysfunction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymph/immunology , Mesentery/immunology , Shock, Hemorrhagic/complications , Systemic Inflammatory Response Syndrome/immunology , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Interleukin-2/metabolism , Interleukin-4/metabolism , Lymph/metabolism , Lymphatic Vessels , Lymphocyte Count , Male , Mesentery/metabolism , Mice , Primary Cell Culture , Receptors, Interleukin-2/metabolism , Shock, Hemorrhagic/blood , Shock, Hemorrhagic/immunology , Systemic Inflammatory Response Syndrome/blood
4.
Semin Immunol ; 28(3): 278-84, 2016 06.
Article in English | MEDLINE | ID: mdl-27157701

ABSTRACT

After severe trauma, the immune system is challenged with a multitude of endogenous and exogenous danger molecules. The recognition of released danger patterns is one of the prime tasks of the innate immune system. In the last two decades, numerous studies have established the complement cascade as a major effector system that detects and processes such danger signals. Animal models with engineered deficiencies in certain complement proteins have demonstrated that widespread complement activation after severe injury culminates in complement dysregulation and excessive generation of complement activation fragments. Such exuberant pro-inflammatory signaling evokes systemic inflammation, causes increased susceptibility to infections and is associated with a detrimental course of the disease after injury. We discuss the underlying processes of such complementopathy and recapitulate different intervention strategies within the complement cascade. So far, several orthogonal anti-complement approaches have been tested with varying success in a large number of rodent, in several porcine and few simian studies. We illustrate the different features among those intervention strategies and highlight those that hold the greatest promise to become potential therapeutic options for the intricate disease of traumatic injury.


Subject(s)
Complement Inactivating Agents/therapeutic use , Immunotherapy/methods , Inflammation/therapy , Shock, Hemorrhagic/therapy , Wounds and Injuries/therapy , Animals , Disease Models, Animal , Humans , Immunity, Innate , Inflammation/immunology , Mice , Mice, Knockout , Pathogen-Associated Molecular Pattern Molecules/immunology , Shock, Hemorrhagic/immunology , Wounds and Injuries/immunology
5.
Cytometry A ; 95(11): 1167-1177, 2019 11.
Article in English | MEDLINE | ID: mdl-31595661

ABSTRACT

Severe injury and hemorrhagic shock (HS) result in multiple changes to hematopoietic differentiation, which contribute to the development of immunosuppression and multiple organ failure (MOF). Understanding the changes that take place during the acute injury phase may help predict which patients will develop MOF and provide potential targets for therapy. Obtaining bone marrow from humans during the acute injury phase is difficult so published data are largely derived from peripheral blood samples, which infer bone marrow changes that reflect the sustained inflammatory response. This preliminary and opportunistic study investigated leucopoietic changes in rat bone marrow 6 h following traumatic injury and HS. Terminally anesthetized male Porton Wistar rats were allocated randomly to receive a sham operation (cannulation with no injury) or femoral fracture and HS. Bone marrow cells were flushed from rat femurs and immunophenotypically stained with specific antibody panels for lymphoid (CD45R, CD127, CD90, and IgM) or myeloid (CD11b, CD45, and RP-1) lineages. Subsequently, cell populations were fluorescence-activated cell sorted for morphological assessment. Stage-specific cell populations were identified using a limited number of antibodies, and leucopoietic changes were determined 6 h following trauma and HS. Myeloid subpopulations could be identified by varying levels CD11b expression, CD45, and RP-1. Trauma and HS resulted in a significant reduction in total CD11b + myeloid cells including both immature (RP-1(-)) and mature (RP-1+) granulocytes. Multiple B-cell lymphoid subsets were identified. The total percentage of CD90+ subsets remained unchanged following trauma and HS, but there was a reduction in the numbers of maturing CD90(-) cells suggesting movement into the periphery. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Subject(s)
Bone Marrow Cells/cytology , Femoral Fractures/immunology , Hematopoietic Stem Cells/cytology , Shock, Hemorrhagic/immunology , Wounds and Injuries/immunology , Animals , Antimicrobial Cationic Peptides/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD11b Antigen/metabolism , Cell Lineage/immunology , Flow Cytometry , Granulocytes/cytology , Granulocytes/metabolism , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Inflammation/immunology , Inflammation/metabolism , Leukocyte Common Antigens/metabolism , Lymphopoiesis/immunology , Male , Multiple Organ Failure/immunology , Multiple Organ Failure/pathology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Rats , Rats, Wistar , Shock, Hemorrhagic/metabolism , Thy-1 Antigens/metabolism , Wounds and Injuries/metabolism
6.
FASEB J ; 32(1): 97-110, 2018 01.
Article in English | MEDLINE | ID: mdl-28855278

ABSTRACT

Acute lung injury (ALI) is a common cause of morbidity in patients after severe injury due to dysregulated inflammation, which is believed to be driven by gut-derived inflammatory mediators carried via mesenteric lymph (ML). We have previously demonstrated that nano-sized extracellular vesicles, called exosomes, secreted into ML after trauma/hemorrhagic shock (T/HS) have the potential to activate immune cells in vitro Here, we assess the function of ML exosomes in the development of T/HS-induced ALI and the role of TLR4 in the ML exosome-mediated inflammatory response. ML exosomes isolated from rats subjected to T/HS stimulated NF-κB activation and caused proinflammatory cytokine production in alveolar macrophages. In vivo experiments revealed that intravenous injection of exosomes harvested after T/HS, but not before shock, caused recruitment of inflammatory cells in the lung, increased vascular permeability, and induced histologic ALI in naive mice. The exosome-depleted supernatant of ML had no effect on in vitro and in vivo inflammatory responses. We also demonstrated that both pharmacologic inhibition and genetic knockout of TLR4 completely abolished ML exosome-induced cytokine production in macrophages. Thus, our findings define the critical role of exosomes secreted into ML as a critical mediator of T/HS-induced ALI through macrophage TLR4 activation.-Kojima, M., Gimenes-Junior, J. A., Chan, T. W., Eliceiri, B. P., Baird, A., Costantini, T. W., Coimbra, R. Exosomes in postshock mesenteric lymph are key mediators of acute lung injury triggering the macrophage activation via Toll-like receptor 4.


Subject(s)
Acute Lung Injury/immunology , Exosomes/microbiology , Macrophage Activation/immunology , Shock, Hemorrhagic/immunology , Toll-Like Receptor 4/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Humans , In Vitro Techniques , Inflammation Mediators/metabolism , Lymph/immunology , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/etiology , Signal Transduction , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/deficiency
7.
Cytokine ; 103: 127-132, 2018 03.
Article in English | MEDLINE | ID: mdl-28969938

ABSTRACT

INTRODUCTION: PPARß/δ agonists are known to modulate the systemic inflammatory response after sepsis. In this study, inflammation modulation effects of PPARß/δ are investigated using the selective PPARß/δ agonist (GW0742) in a model of haemorrhagic shock (HS)-induced sterile systemic inflammation. METHODS: Blood pressure-controlled (35±5mmHg) HS was performed in C57/BL6 mice for 90min. Low-dose GW0742 (0.03mg/kg/BW) and high-dose GW0742 (0.3mg/kg/BW) were then administered at the beginning of resuscitation. Mice were sacrificed 6h after induction of HS. Plasma levels of IL-6, IL-1ß, IL-10, TNFα, KC, MCP-1, and GM-CSF were determined by ELISA. Myeloperoxidase (MPO) activity in pulmonary and liver tissues was analysed with standardised MPO kits. RESULTS: In mice treated with high-dose GW0742, plasma levels of IL-6, IL-1ß, and MCP-1 were significantly increased compared to the control group mice. When compared to mice treated with low-dose GW0742 plasma levels of IL-6, IL-1ß, GM-CSF, KC, and MCP-1 were significantly elevated in high-dose-treated mice. Low-dose GW0742 treatment was associated with a non-significant downtrend of inflammatory factors in mice with HS. No significant changes of MPO activity in lung and liver were observed between the control group and the GW0742 treatment groups. CONCLUSION: This study identified dose-dependent effects of GW0742 on systemic inflammation after HS. While high-dose GW0742 substantially enhanced the systemic inflammatory response, low-dose GW0742 led to a downtrend of pro-inflammation cytokine expression. The exact mechanisms are yet unknown and need to be assessed in further studies.


Subject(s)
PPAR delta/agonists , PPAR-beta/agonists , Shock, Hemorrhagic/drug therapy , Systemic Inflammatory Response Syndrome/drug therapy , Thiazoles/pharmacology , Animals , Cytokines/immunology , Dose-Response Relationship, Drug , Male , Mice , PPAR delta/immunology , PPAR-beta/immunology , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/immunology , Systemic Inflammatory Response Syndrome/etiology , Systemic Inflammatory Response Syndrome/immunology
8.
J Surg Res ; 229: 262-270, 2018 09.
Article in English | MEDLINE | ID: mdl-29936999

ABSTRACT

BACKGROUND: Post-traumatic lung injury following trauma and hemorrhagic shock (T/HS) is associated with significant morbidity. Leukotriene-induced inflammation has been implicated in the development of post-traumatic lung injury through a mechanism that is only partially understood. Postshock mesenteric lymph returning to the systemic circulation is rich in arachidonic acid, the substrate of 5-lipoxygenase (ALOX5). ALOX5 is the rate-limiting enzyme in leukotriene synthesis and, following T/HS, contributes to the development of lung dysfunction. ALOX5 colocalizes with its cofactor, 5-lipoxygenase-activating protein (ALOX5AP), which is thought to potentiate ALOX5 synthetic activity. We hypothesized that T/HS results in the molecular association and nuclear colocalization of ALOX5 and ALOX5AP, which ultimately increases leukotriene production and potentiates lung injury. MATERIALS AND METHODS: To examine these molecular interactions, a rat T/HS model was used. Post-T/HS tissue was evaluated for lung injury through both histologic analysis of lung sections and biochemical analysis of bronchoalveolar lavage fluid. Lung tissue was immunostained for ALOX5 and ALOX5AP with association and colocalization evaluated by fluorescence resonance energy transfer. In addition, rats undergoing T/HS were treated with MK-886, a known ALOX5AP inhibitor. RESULTS: ALOX5 levels increase and ALOX5/ALOX5AP association occurred after T/HS, as evidenced by increases in total tissue fluorescence and fluorescence resonance energy transfer signal intensity, respectively. These findings coincided with increased leukotriene production and with the histological changes characteristic of lung injury. ALOX5/ALOX5AP complex formation, leukotriene production, and lung injury were decreased after inhibition of ALOX5AP with MK-886. CONCLUSIONS: These results suggest that the association of ALOX5/ALOX5AP contributes to leukotriene-induced inflammation and predisposes the T/HS animal to lung injury.


Subject(s)
5-Lipoxygenase-Activating Proteins/immunology , Acute Lung Injury/immunology , Arachidonate 5-Lipoxygenase/immunology , Shock, Hemorrhagic/immunology , 5-Lipoxygenase-Activating Proteins/metabolism , Acute Lung Injury/pathology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Disease Models, Animal , Humans , Leukotrienes/immunology , Leukotrienes/metabolism , Lung/immunology , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/immunology , Shock, Hemorrhagic/pathology
9.
Gen Physiol Biophys ; 37(1): 83-92, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29424353

ABSTRACT

Hemorrhagic shock (HS) still has a high mortality rate and none of the known resuscitative regimens completely reverse its adverse outcomes. This study investigated the effects of different models of resuscitative therapy on the healing of organ damage in a HS model. Male Wistar rats were randomized into six groups: Sham, without HS induction; HS, without resuscitation; HS+Blood, resuscitation with the shed blood; HS+Blood+NS, resuscitation with blood and normal saline; HS+Blood+RL, resuscitation with blood and Ringer's lactate; EPO, erythropoietin was added to the blood and RL. Blood and urine samples were obtained 3 h after resuscitation. Kidney, liver and brain tissue samples were harvested for multiple organ failure evaluation. Survival rate was the highest in the Sham, EPO and HS+Blood+RL groups compared to others. Plasma creatinine concentration, ALT, AST, urinary NAG activity and renal NGAL mRNA expression significantly increased in the HS+Blood+RL group compared to the Sham group. There was a significant increase in tissue oxidative stress markers and pro-inflammatory cytokines in HS+Blood+RL group compared to the Sham rats. EPO had more protective effects on multiple organ failure compared to the HS+Blood+RL group. EPO, as a resuscitative treatment, attenuated HS-induced organ damage. It seems that it has a potential to be attractive for clinical trials.


Subject(s)
Erythropoietin/administration & dosage , Models, Animal , Multiple Organ Failure/drug therapy , Oxidative Stress/immunology , Resuscitation/methods , Shock, Hemorrhagic/drug therapy , Shock, Hemorrhagic/immunology , Animals , Anti-Inflammatory Agents , Critical Illness/therapy , Dose-Response Relationship, Drug , Male , Multiple Organ Failure/immunology , Multiple Organ Failure/pathology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Severity of Illness Index , Shock, Hemorrhagic/pathology , Treatment Outcome , Viscera/drug effects , Viscera/immunology
10.
PLoS Med ; 14(7): e1002365, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742815

ABSTRACT

BACKGROUND: The immunosuppression and immune dysregulation that follows severe injury includes type 2 immune responses manifested by elevations in interleukin (IL) 4, IL5, and IL13 early after injury. We hypothesized that IL33, an alarmin released early after tissue injury and a known regulator of type 2 immunity, contributes to the early type 2 immune responses after systemic injury. METHODS AND FINDINGS: Blunt trauma patients admitted to the trauma intensive care unit of a level I trauma center were enrolled in an observational study that included frequent blood sampling. Dynamic changes in IL33 and soluble suppression of tumorigenicity 2 (sST2) levels were measured in the plasma and correlated with levels of the type 2 cytokines and nosocomial infection. Based on the observations in humans, mechanistic experiments were designed in a mouse model of resuscitated hemorrhagic shock and tissue trauma (HS/T). These experiments utilized wild-type C57BL/6 mice, IL33-/- mice, B6.C3(Cg)-Rorasg/sg mice deficient in group 2 innate lymphoid cells (ILC2), and C57BL/6 wild-type mice treated with anti-IL5 antibody. Severely injured human blunt trauma patients (n = 472, average injury severity score [ISS] = 20.2) exhibited elevations in plasma IL33 levels upon admission and over time that correlated positively with increases in IL4, IL5, and IL13 (P < 0.0001). sST2 levels also increased after injury but in a delayed manner compared with IL33. The increases in IL33 and sST2 were significantly greater in patients that developed nosocomial infection and organ dysfunction than similarly injured patients that did not (P < 0.05). Mechanistic studies were carried out in a mouse model of HS/T that recapitulated the early increase in IL33 and delayed increase in sST2 in the plasma (P < 0.005). These studies identified a pathway where IL33 induces ILC2 activation in the lung within hours of HS/T. ILC2 IL5 up-regulation induces further IL5 expression by CXCR2+ lung neutrophils, culminating in early lung injury. The major limitations of this study are the descriptive nature of the human study component and the impact of the potential differences between human and mouse immune responses to polytrauma. Also, the studies performed did not permit us to make conclusions about the impact of IL33 on pulmonary function. CONCLUSIONS: These results suggest that IL33 may initiate early detrimental type 2 immune responses after trauma through ILC2 regulation of neutrophil IL5 production. This IL33-ILC2-IL5-neutrophil axis defines a novel regulatory role for ILC2 in acute lung injury that could be targeted in trauma patients prone to early lung dysfunction.


Subject(s)
Gene Expression Regulation , Immunity, Humoral , Interleukin-33/metabolism , Interleukin-5/genetics , Lymphocytes/immunology , Wounds and Injuries/immunology , Adult , Aged , Aged, 80 and over , Animals , Cohort Studies , Disease Models, Animal , Female , Humans , Interleukin-33/blood , Interleukin-5/immunology , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Retrospective Studies , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/immunology , Wounds and Injuries/etiology , Wounds and Injuries/genetics , Young Adult
11.
J Immunol ; 195(5): 2396-407, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26246141

ABSTRACT

The elderly are particularly susceptible to trauma, and their outcomes are frequently dismal. Such patients often have complicated clinical courses and ultimately die of infection and sepsis. Recent research has revealed that although elderly subjects have increased baseline inflammation as compared with their younger counterparts, the elderly do not respond to severe infection or injury with an exaggerated inflammatory response. Initial retrospective analysis of clinical data from the Glue Grant trauma database demonstrated that despite a similar frequency, elderly trauma patients have worse outcomes to pneumonia than younger subjects do. Subsequent analysis with a murine trauma model also demonstrated that elderly mice had increased mortality after posttrauma Pseudomonas pneumonia. Blood, bone marrow, and bronchoalveolar lavage sample analyses from juvenile and 20-24-mo-old mice showed that increased mortality to trauma combined with secondary infection in the aged are not due to an exaggerated inflammatory response. Rather, they are due to a failure of bone marrow progenitors, blood neutrophils, and bronchoalveolar lavage cells to initiate and complete an emergency myelopoietic response, engendering myeloid cells that fail to clear secondary infection. In addition, elderly people appeared unable to resolve their inflammatory response to severe injury effectively.


Subject(s)
Aging/immunology , Immunity/immunology , Myelopoiesis/immunology , Shock, Hemorrhagic/immunology , Wounds and Injuries/immunology , Adult , Age Factors , Aged , Aging/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Cohort Studies , Female , Humans , Immunity/genetics , Leukocytes/immunology , Leukocytes/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Myelopoiesis/genetics , Oligonucleotide Array Sequence Analysis , Pneumonia, Ventilator-Associated/etiology , Pneumonia, Ventilator-Associated/immunology , Pseudomonas Infections/genetics , Pseudomonas Infections/immunology , Pseudomonas Infections/mortality , Shock, Hemorrhagic/complications , Survival Rate , Transcriptome/genetics , Transcriptome/immunology , Wounds and Injuries/complications
12.
Chin J Traumatol ; 20(4): 207-211, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28684036

ABSTRACT

PURPOSE: It has been suggested that patients with traumatic insults are resuscitated into a state of an early systemic inflammatory response. We aimed to evaluate the influence of hemorrhagic shock and resuscitation (HSR) upon the inflammatory response capacity assessed by overall TNF-α secretion capacity of the host compared to its release from circulating leukocytes in peripheral circulation. METHODS: Rats (8/group) subjected to HS (MAP of 30-35 mmHg for 90 min followed by resuscitation over 50 min) were challenged with Lipopolysaccharide (LPS), 1 µg/kg intravenously at the end of resuscitation (HSR-LPS group) or 24 h later (HSR-LPS24 group). Control animals were injected with LPS without bleeding (LPS group). Plasma TNF-α was measured at 90 min after the LPS challenge. In addition, whole blood (WB) was obtained either from healthy controls (CON) immediately after resuscitation (HSR), or at 24 h post-shock (HSR 24). WB was incubated with LPS (100 ng/mL) for 2 h at 37 °C. TNF-α concentration and LPS binding capacity (LBC) was determined. RESULTS: Compared to LPS group, HSR followed by LPS challenge resulted in suppression of plasma TNF-α in HSR-LPS and HSR-LPS24 groups (1835 ± 478, 273 ± 77, 498 ± 200 pg/mL, respectively). Compared to CON the LPS-induced TNF-α release capacity of circulating leukocytes ex vivo was strongly declined both at the end of resuscitation (HSR) and 24 h later (HSR24) (1012 ± 259, 313 ± 154, 177 ± 63 ng TNF/mL, respectively). The LBC in WB was similar between CON and HSR and only moderately enhanced in HSR24 (57 ± 6, 56 ± 6, 71 ± 5 %, respectively). CONCLUSION: Our data suggest that the overall inflammatory response capacity is decreased immediately after HSR, persisting up to 24 h, and is independent of LBC.


Subject(s)
Resuscitation , Shock, Hemorrhagic/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood
13.
Genet Mol Res ; 15(1)2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26909947

ABSTRACT

The aim of this study was to explore the relationship between adiponectin (ADPN) and hemorrhagic shock (HS) and the recovery after HS. This is significant for further understanding of the pathophysiological processes of HS and the development of better treatments. In total, 72 male C57BL/6 mice were assigned randomly to three groups: control, HS, and recovery (N = 24). The HS mouse model was constructed by hemorrhage of the carotid artery and recovery was achieved by tail vein injection of Ringer's solution. The level of ADPN in the peripheral blood of mice before and after recovery was detected by enzyme-linked immunosorbent assay. Compared to control, HS mice showed significantly decreased ADPN levels with the extension of HS time while the level of ADPN in recovery mice increased significantly and remained high. The variation of ADPN levels was closely associated with the occurrence of HS in mice and their recovery, suggesting that ADPN might act as a biomarker of inflammation and have potential for the treatment of HS.


Subject(s)
Adiponectin/blood , Disease Models, Animal , Inflammation , Recovery of Function , Shock, Hemorrhagic/blood , Adiponectin/immunology , Animals , Biomarkers , Carotid Arteries/metabolism , Carotid Arteries/physiopathology , Male , Mice , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/physiopathology
14.
Am J Physiol Gastrointest Liver Physiol ; 309(3): G202-8, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26045612

ABSTRACT

CD103(+) dendritic cells (DCs) continuously migrate from the intestine to the mesenteric lymph nodes (MLNs) and maintain tolerance by driving the development of regulatory T cells (Treg) in the gut. The relative expression of Treg and T-helper 17 (Th17) cells determines the balance between tolerance and immunity in the gut. We hypothesized that trauma/hemorrhagic shock (T/HS) would decrease the CD103(+) DC population in the mesenteric lymph and alter the Treg-to-Th17 ratio in the MLN. We further hypothesized that vagus nerve stimulation (VNS) would promote tolerance to inflammation by increasing the Treg-to-Th17 ratio in the MLN after injury. Male rats were assigned to sham shock (SS), trauma/sham shock (T/SS), or T/HS. T/HS was induced by laparotomy and 60 min of HS (blood pressure 35 mmHg) followed by fluid resuscitation. A separate cohort of animals underwent cervical VNS after the HS phase. MLN samples were collected 24 h after resuscitation. The CD103(+) DC population and Treg-to-Th17 cell ratio in the MLN were decreased after T/HS compared with SS and T/SS, suggesting a shift to an inflammatory response. VNS prevented the T/HS-induced decrease in the CD103(+) DC population and increased the Treg-to-Th17 ratio compared with T/HS alone. VNS alters the gut inflammatory response to injury by modulating the Treg-Th17 cell balance in the MLN. VNS promotes tolerance to inflammation in the gut, further supporting its ability to modulate the inflammatory set point and alter the response to injury.


Subject(s)
Dendritic Cells/immunology , Inflammation/immunology , Lymph Nodes/metabolism , Mesentery , Shock, Hemorrhagic/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antigens, CD/immunology , Cell Movement/physiology , Disease Models, Animal , Inflammation/etiology , Integrin alpha Chains/immunology , Intestines/immunology , Male , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/complications , Vagus Nerve Stimulation
15.
Ann Surg ; 261(6): 1215-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25185480

ABSTRACT

OBJECTIVE: To evaluate the efficacy of remote ischemic conditioning (RIC) on organ protection after hemorrhagic shock/resuscitation (S/R) in a murine model. BACKGROUND: Ischemia/reperfusion resulting from S/R contributes to multiple organ dysfunction in trauma patients. We hypothesized that RIC before shock (remote ischemic preconditioning), during shock (remote ischemic "PER"conditioning), or during resuscitation (remote ischemic "POST"conditioning) could confer organ protection. We also tested the effect of ischemic conditioned plasma on neutrophil migration in vivo using transgenic zebrafish models. METHODS: C57Bl/6 mice were subjected to S/R with or without hindlimb RIC. Serum levels of alanine aminotransferase and tumor necrosis factor-alpha, and liver tumor necrosis factor-alpha and interleukin 1ß mRNA were evaluated. In some experiments, lung protein leakage, cytokines, and myeloperoxidase activity were investigated. Plasma from mice subjected to RIC was microinjected into zebrafish, and neutrophil migration was assessed after tailfin transection or copper sulfate treatment. RESULTS: In mice subjected to S/R, remote ischemic preconditioning, remote ischemic "PER"conditioning, and remote ischemic "POST"conditioning each significantly reduced serum alanine aminotransferase and liver mRNA expression of tumor necrosis factor-alpha and interleukin 1ß and improved liver histology compared with control S/R mice. Lung injury and inflammation were also significantly reduced in mice treated with remote ischemic preconditioning. Zebrafish injected with plasma or dialyzed plasma (fraction >14 kDa) from ischemic conditioned mice had reduced neutrophil migration toward sites of injury compared with zebrafish injected with control plasma. CONCLUSIONS: RIC protects against S/R-induced organ injury, in part, through a humoral factor(s), which alters neutrophil function. The beneficial effects of RIC, performed during the S/R phase of care, suggest a role for its application early in the posttrauma period.


Subject(s)
Ischemic Preconditioning , Liver Diseases/blood , Lung Injury/blood , Reperfusion Injury/blood , Shock, Hemorrhagic/blood , Shock, Hemorrhagic/therapy , Alanine Transaminase/blood , Animals , Animals, Genetically Modified , Biomarkers/blood , Chemotaxis, Leukocyte , Disease Models, Animal , Interleukin-1beta/blood , Liver Diseases/etiology , Liver Diseases/pathology , Lung Injury/etiology , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Plasma/immunology , Reperfusion Injury/etiology , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/prevention & control , Time Factors , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Zebrafish
16.
J Surg Res ; 194(1): 220-32, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25481528

ABSTRACT

Acute traumatic injury is a complex disease that has remained a leading cause of death, which affects all ages in our society. Direct mechanical insult to tissues may result in physiological and immunologic disturbances brought about by blood loss, coagulopathy, as well as ischemia and reperfusion insults. This inappropriate response leads to an abnormal release of endogenous mediators of inflammation that synergistically contribute to the incidence of morbidity and mortality. This aberrant activation and suppression of the immune system follows a bimodal pattern, wherein activation of the innate immune responses is followed by an anti-inflammatory response with suppression of the adaptive immunity, which can subsequently lead secondary insults and multiple organ dysfunction. Traumatic injury rodent and swine models have been used to describe many of the underlying pathologic mechanisms, which have led to an improved understanding of the morbidity and mortality associated with critically ill trauma patients. The enigmatic immunopathology of the human immunologic response after severe trauma, however, has never more been apparent and there grows a need for a clinically relevant animal model, which mimics this immune physiology to enhance the care of the most severely injured. This has necessitated preclinical studies in a more closely related model system, the nonhuman primate. In this review article, we summarize animal models of trauma that have provided insight into the clinical response and understanding of cellular mechanisms involved in the onset and progression of ischemia-reperfusion injury as well as describe future treatment options using immunomodulation-based strategies.


Subject(s)
Disease Models, Animal , Wounds and Injuries/immunology , Acute Disease , Animals , Complement Activation , Cytokines/physiology , Humans , Neutrophils/physiology , Shock, Hemorrhagic/immunology
17.
J Surg Res ; 193(1): 421-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25224275

ABSTRACT

BACKGROUND: This study investigated the effects of pentoxifylline (PTX) combined with resuscitation fluids on microcirculatory dysfunctions in a two-hit model of shock and sepsis. MATERIALS AND METHODS: Male Wistar rats (250 g) were submitted to hemorrhagic shock and reperfusion followed by sepsis induced by cecal ligation and puncture. For the initial treatment of shock, rats were randomly divided into: sham, no injury, no treatment; hypertonic saline solution (HS) (7.5%, 4 mL/kg); lactated Ringer's solution (LR, 3 × shed blood volume); HS + PTX (4 mL/Kg + 25 mg/kg PTX); and LR + PTX (3 × shed blood volume + 25 mg/kg PTX). After 48 h of being exposed to the double injury, leukocyte-endothelial interactions were assessed by intravital microscopy of the mesentery. Endothelial expression of P-selectin and intercellular adhesion molecule-1 (ICAM-1) was evaluated by immunohistochemistry, as well as lung neutrophil infiltration by histology. RESULTS: Lactated Ringer's solution induced marked increases (P < 0.001) in the number of rolling leukocytes per 10 min (two-fold), adherent leukocytes per 100 µm venule length (six-fold), migrated leukocytes per 5000 µm(2) (eight-fold), P-selectin and ICAM-1 expression (four-fold), and lung neutrophil infiltration (three-fold) compared with sham. In contrast, PTX attenuated leukocyte-endothelial interactions, P-selectin and ICAM-1 expression at the mesentery when associated with either LR (P < 0.001) or HS (P < 0.05). Neutrophil migration into the lungs was similarly reduced by PTX (P < 0.05). CONCLUSIONS: Data presented showed that pentoxifylline attenuates microcirculatory disturbances at the mesenteric bed with significant minimization of lung inflammation after a double-injury model of hemorrhagic shock and reperfusion followed by sepsis.


Subject(s)
Cell Communication/drug effects , Endothelial Cells/drug effects , Leukocyte Rolling/drug effects , Pentoxifylline/pharmacology , Sepsis/drug therapy , Shock, Hemorrhagic/drug therapy , Animals , Cecum/injuries , Disease Models, Animal , Free Radical Scavengers/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Isotonic Solutions/pharmacology , Ligation , Male , Microcirculation/drug effects , Multiple Organ Failure/drug therapy , Multiple Organ Failure/immunology , P-Selectin/metabolism , Rats, Wistar , Resuscitation/methods , Ringer's Lactate , Sepsis/immunology , Shock, Hemorrhagic/immunology , Wounds, Stab
18.
J Immunol ; 190(10): 5247-55, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23585683

ABSTRACT

Hemorrhagic shock (HS) promotes the development of systemic inflammatory response syndrome and organ injury by activating and priming the innate immune system for an exaggerated inflammatory response through, as of yet, unclear mechanisms. IL-1ß also plays an important role in the development of post-HS systemic inflammatory response syndrome and active IL-1ß production is tightly controlled by the inflammasome. Pyrin, a protein of 781 aa with pyrin domain at the N-terminal, negatively regulates inflammasome activation through interaction with nucleotide-binding oligomerization domain-like receptor protein (NLRP). Expression of pyrin can be induced by LPS and cytokines, and IL-10 is a known potent inducer of pyrin expression in macrophages. In the current study, we tested the hypothesis that HS downregulates IL-10 and therefore decreases pyrin expression to promote inflammasome activation and subsequent IL-1ß processing and secretion in the lungs. Our results show that LPS, while activating Nlrp3 inflammasome in the lungs, also induced pyrin expression, which in turn suppressed inflammasome activation. More importantly, LPS-mediated upregulation of IL-10 enhanced pyrin expression, which serves, particularly in later phases, as a potent negative-feedback mechanism regulating inflammasome activation. However, HS-mediated suppression of IL-10 expression in alveolar macrophages attenuated the upregulation of pyrin in alveolar macrophages and lung endothelial cells and thereby significantly enhanced inflammasome activation and IL-1ß secretion in the lungs. This study demonstrates a novel mechanism by which HS suppresses negative-feedback regulation of Nlrp3 inflammasome to enhance IL-1ß secretion in response to subsequent LPS challenge and so primes for inflammation.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Inflammasomes/immunology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Shock, Hemorrhagic/immunology , Animals , Carrier Proteins/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Disease Models, Animal , Down-Regulation , Endothelial Cells/immunology , Endothelial Cells/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/biosynthesis , Interleukin-1beta/immunology , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Pyrin , RNA Interference , RNA, Small Interfering , Signal Transduction/immunology , Systemic Inflammatory Response Syndrome , Toll-Like Receptor 4/genetics , Up-Regulation
19.
J Surg Res ; 191(2): 423-31, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24836421

ABSTRACT

BACKGROUND: Resuscitative endovascular balloon occlusion of the aorta (REBOA) is a hemorrhage control and resuscitative adjunct that has been demonstrated to improve central perfusion during hemorrhagic shock. The aim of this study was to characterize the systemic inflammatory response associated and cardiopulmonary sequelae with 30, 60, and 90 min of balloon occlusion and shock on the release of interleukin 6 (IL-6) and tumor necrosis factor alpha. MATERIALS AND METHODS: Anesthetized female Yorkshire swine (Sus scrofa, weight 70-90 kg) underwent a 35% blood volume-controlled hemorrhage followed by thoracic aortic balloon occlusion of 30 (30-REBOA, n = 6), 60 (60-REBOA, n = 8), and 90 min (90-REBOA, n = 6). This was followed by resuscitation with whole blood and crystalloid over 6 h. Animals then underwent 48 h of critical care with sedation, fluid, and vasopressor support. RESULTS: All animals were successfully induced into hemorrhagic shock without mortality. All groups responded to aortic occlusion with a rise in blood pressure above baseline values. IL-6, as measured (picogram per milliliter) at 8 h, was significantly elevated from baseline values in the 60-REBOA and 90-REBOA groups: 289 ± 258 versus 10 ± 5; P = 0.018 and 630 ± 348; P = 0.007, respectively. There was a trend toward greater vasopressor use (P = 0.183) and increased incidence of acute respiratory distress syndrome (P = 0.052) across the groups. CONCLUSIONS: REBOA is a useful adjunct in supporting central perfusion during hemorrhagic shock; however, increasing occlusion time and shock results in a greater IL-6 release. Clinicians must anticipate inflammation-mediated organ failure in post-REBOA use patients.


Subject(s)
Balloon Occlusion/adverse effects , Inflammation/etiology , Shock, Hemorrhagic/therapy , Animals , Aorta , Female , Hemodynamics , Interleukin-6/blood , Resuscitation , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/physiopathology , Swine , Tumor Necrosis Factor-alpha/blood
20.
Mil Med ; 189(Supplement_3): 416-422, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160884

ABSTRACT

INTRODUCTION: Many trauma patients die from hemorrhagic shock in the military and civilian settings. Although two-thirds of hemorrhagic shock victims die of reasons other than exsanguination, such as the consequent cytokine storm, anti-inflammatory therapies failed to be utilized. Apoptotic cell-based treatments enhance innate ability to exert systemic immunomodulation as demonstrated in several clinical applications and hence might present a novel approach in hemorrhagic shock treatment. MATERIALS AND METHODS: Twenty-two rats underwent a pressure-controlled hemorrhagic shock model and followed up for 24 hours. An infusion of apoptotic cells (Allocetra-OTS, Enlivex Therapeutics Ltd, Nes Ziona, Israel) was administered to the treatment group. Hemodynamics, blood counts, biochemistry findings, and cytokine profile were compared to a saline-resuscitated control group. RESULTS: The treatment group's mean arterial pressure decreased from 94.8 mmHg to 28.2 mmHg, resulting in an 8.13 mg/dL increase in lactate and a 1.9 g/L decrease in hemoglobin, similar to the control group. White blood cells and platelets decreased more profoundly in the treatment group. A similar cytokine profile after 24 hours was markedly attenuated in the treatment group 2 hours after bleeding. Levels of pro-inflammatory cytokines such as interleukin (IL)-1a (28.4 pg/mL vs. 179.1 pg/mL), IL-1b (47.4 pg/mL vs. 103.9 pg/mL), IL-6 (526.2 pg/mL vs. 3492 pg/mL), interferon γ (11.4 pg/mL vs. 427.9 pg/mL), and tumor necrosis factor α (19.0 pg/mL vs. 31.7 pg/mL) were profoundly lower in the treatment group. CONCLUSION: In a pressure-control hemorrhagic shock model in rats, apoptotic cell infusion showed preliminary signs of a uniform attenuated cytokine response. Apoptotic cell-based therapies might serve as a novel immunomodulatory therapy for hemorrhagic shock.


Subject(s)
Apoptosis , Shock, Hemorrhagic , Shock, Hemorrhagic/therapy , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/immunology , Animals , Rats , Male , Apoptosis/physiology , Rats, Sprague-Dawley , Disease Models, Animal , Cell- and Tissue-Based Therapy/methods , Cytokines/blood , Cytokines/analysis , Inflammation/therapy
SELECTION OF CITATIONS
SEARCH DETAIL