Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Cells ; 10(7)2021 06 30.
Article in English | MEDLINE | ID: mdl-34209240

ABSTRACT

We previously reported an early surge in high mobility group box protein 1 (HMGB1) levels in a polytrauma (PT) rat model. This study investigates the association of HMGB1 levels in mediating PT associated dysregulated immune responses and its influence on the cellular levels of receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4). Using the same PT rat model treated with anti-HMGB1 polyclonal antibody, we evaluated changes in circulating inflammatory cytokines, monocytes/macrophages and T cells dynamics and cell surface expression of RAGE and TLR4 at 1, 3, and 7 days post-trauma (dpt) in blood and spleen. Notably, PT rats demonstrating T helper (Th)1 and Th2 cells type early hyper-inflammatory responses also exhibited increased monocyte/macrophage counts and diminished T cell counts in blood and spleen. In blood, expression of RAGE and TLR4 receptors was elevated on CD68+ monocyte/macrophages and severely diminished on CD4+ and CD8+ T cells. Neutralization of HMGB1 significantly decreased CD68+ monocyte/macrophage counts and increased CD4+ and CD8+ T cells, but not γδ+TCR T cells in circulation. Most importantly, RAGE and TLR4 expressions were restored on CD4+ and CD8+ T cells in treated PT rats. Overall, findings suggest that in PT, the HMGB1 surge is responsible for the onset of T cell exhaustion and dysfunction, leading to diminished RAGE and TLR4 surface expression, thereby possibly hindering the proper functioning of T cells.


Subject(s)
HMGB1 Protein/metabolism , Inflammation/immunology , Multiple Trauma/immunology , T-Lymphocytes/immunology , Animals , Antibodies/pharmacology , Blood Proteins/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Count , Cell Membrane/metabolism , Cytokines/blood , Disease Models, Animal , Male , Models, Biological , Multiple Trauma/blood , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neutralization Tests , Osteotomy , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/metabolism , Spleen/immunology , Toll-Like Receptor 4/metabolism
2.
Shock ; 56(5): 658-666, 2021 11 01.
Article in English | MEDLINE | ID: mdl-33882515

ABSTRACT

ABSTRACT: Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells hallmarked by their potent immunosuppressive function in a vast array of pathologic conditions. MDSCs have recently been shown to exhibit marked expansion in acute inflammatory states including traumatic injury, burn, and sepsis. Although MDSCs have been well characterized in cancer, there are significant gaps in our knowledge of their functionality in trauma and sepsis, and their clinical significance remains unclear. It is suggested that MDSCs serve an important role in quelling profound inflammatory responses in the acute setting; however, MDSC accumulation may also predispose patients to developing persistent immune dysregulation with increased risk for nosocomial infections, sepsis, and multiorgan failure. Whether MDSCs may serve as the target for novel therapeutics or an important biomarker in trauma and sepsis is yet to be determined. In this review, we will discuss the current understanding of MDSCs within the context of specific traumatic injury types and sepsis. To improve delineation of their functional role, we propose a systemic approach to MDSC analysis including phenotypic standardization, longitudinal analysis, and expansion of clinical research.


Subject(s)
Inflammation/immunology , Myeloid-Derived Suppressor Cells/physiology , Wounds and Injuries/immunology , Humans
3.
Cytokine ; 141: 155457, 2021 05.
Article in English | MEDLINE | ID: mdl-33581471

ABSTRACT

INTRODUCTION: Traumatic injury with hemorrhage (TH) induces an inflammatory response in the lung resulting in lung injury involving activation of immune cells including myeloid cells (i.e., monocytes, granulocytes and macrophages), in part through TLRs. TLRs, via the recognition of damage associated molecular patterns (DAMPs), are a key link between tissue injury and inflammation. Nonetheless, the role of TLRs in myeloid cell activation and TH-induced lung injury remains ill defined. METHODS: C57BL/6 male mice were subjected to TH or sham treatment (n = 4-6 /group). Lung tissues were collected two hrs. after injury. Single cells were isolated from the lungs by enzymatic digestion and myeloid cell TLR expression and activation (i.e., cytokine production) were assessed using flow cytometry techniques. RESULTS: The injury was associated with a profound change in the lung myeloid cell population. TH markedly increased lung CD11b+ monocyte numbers and Gr1+ granulocyte numbers as compared to sham mice. The number of cells expressing TLR2, TLR4, and TLR9 were increased 4-7 fold in the TH mice. Activation for elevated cytokine (TNFα, IL-10) production was observed in the lung monocyte population of the TH mice. CONCLUSIONS: Trauma-induced lung injury is associated with infiltration of the lungs with TLR expressing myeloid cells that are activated for elevated cytokine responses. This elevation in TLR expression may contribute to DAMP-mediated pulmonary complications of an inflammatory nature and warrants further investigation.


Subject(s)
Hemorrhage/immunology , Lung Injury/immunology , Lung/immunology , Myeloid Cells/immunology , Toll-Like Receptors/immunology , Wounds and Injuries/immunology , Animals , Hemorrhage/complications , Interleukin-10/immunology , Lung Injury/etiology , Male , Mice , Tumor Necrosis Factor-alpha/immunology , Wounds and Injuries/complications
4.
J Trauma Acute Care Surg ; 88(5): 579-587, 2020 05.
Article in English | MEDLINE | ID: mdl-32039976

ABSTRACT

BACKGROUND: Traumatic injury can lead to a compromised intestinal epithelial barrier, decreased gut perfusion, and inflammation. While recent studies indicate that the gut microbiome (GM) is altered early following traumatic injury, the impact of GM changes on clinical outcomes remains unknown. Our objective of this follow-up study was to determine if the GM is associated with clinical outcomes in critically injured patients. METHODS: We conducted a prospective, observational study in adult patients (N = 67) sustaining severe injury admitted to a level I trauma center. Fecal specimens were collected on admission to the emergency department, and microbial DNA from all samples was analyzed using the Quantitative Insights Into Microbial Ecology pipeline and compared against the Greengenes database. α-Diversity and ß-diversity were estimated using the observed species metrics and analyzed with t tests and permutational analysis of variance for overall significance, with post hoc pairwise analyses. RESULTS: Our patient population consisted of 63% males with a mean age of 44 years. Seventy-eight percent of the patients suffered blunt trauma with 22% undergoing penetrating injuries. The mean body mass index was 26.9 kg/m. Significant differences in admission ß-diversity were noted by hospital length of stay, intensive care unit hospital length of stay, number of days on the ventilator, infections, and acute respiratory distress syndrome (p < 0.05). ß-Diversity on admission differed in patients who died compared with patients who lived (mean time to death, 8 days). There were also significantly less operational taxonomic units in samples from patients who died versus those who survived. A number of species were enriched in the GM of injured patients who died, which included some traditionally probiotic species such as Akkermansia muciniphilia, Oxalobacter formigenes, and Eubacterium biforme (p < 0.05). CONCLUSION: Gut microbiome diversity on admission in severely injured patients is predictive of a variety of clinically important outcomes. While our study does not address causality, the GM of trauma patients may provide valuable diagnostic and therapeutic targets for the care of injured patients. LEVEL OF EVIDENCE: Prognostic and epidemiological, level III.


Subject(s)
Gastrointestinal Microbiome/physiology , Wounds, Nonpenetrating/mortality , Wounds, Penetrating/mortality , Adult , Aged , Emergency Service, Hospital/statistics & numerical data , Feces/microbiology , Female , Follow-Up Studies , Hospital Mortality , Humans , Injury Severity Score , Length of Stay/statistics & numerical data , Male , Middle Aged , Prognosis , Prospective Studies , Trauma Centers/statistics & numerical data , Wounds, Nonpenetrating/diagnosis , Wounds, Nonpenetrating/microbiology , Wounds, Penetrating/diagnosis , Wounds, Penetrating/microbiology
5.
Cell Immunol ; 338: 21-26, 2019 04.
Article in English | MEDLINE | ID: mdl-30902343

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) have been identified in the burn wound, however their characterization is incomplete. To study this, mice were subjected to a major burn and skin cells were isolated 3 days thereafter for analysis. Significant infiltration of the burn wound with MDSCs was observed as compared with uninjured skin. The skin of naïve mice did not contain MDSCs. Characterization of the cells showed that 33% of MDSCs in the wound were monocytic (M)-MDSCs, which was significantly less than that found in uninjured skin (52%). In contrast, polymorphonuclear (PMN)-MDSCs were greater in the burn wound as compared with uninjured skin. Burn wound TLR expression by both MDSCs subsets was decreased as compared with uninjured skin. Wound MDSCs produced pro- and anti-inflammatory cytokines and iNOS was present in both MDSC subsets, whereas ARG1 was only present in M-MDSCs. In conclusion, both M- and PMN-MDSCs infiltrate burn wound with after injury, however, they displayed decreased TLR expression, suggesting receptor down-regulation.


Subject(s)
Burns/immunology , Monocytes/physiology , Myeloid-Derived Suppressor Cells/physiology , Neutrophils/physiology , Skin/pathology , Animals , Arginase/metabolism , Cell Movement , Cytokines/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Skin/injuries
6.
J Trauma Acute Care Surg ; 86(4): 573-582, 2019 04.
Article in English | MEDLINE | ID: mdl-30633104

ABSTRACT

BACKGROUND: Traumatic injury can lead to a compromised intestinal epithelial barrier and inflammation. While alterations in the gut microbiome of critically injured patients may influence clinical outcomes, the impact of trauma on gut microbial composition is unknown. Our objective was to determine if the gut microbiome is altered in severely injured patients and begin to characterize changes in the gut microbiome due to time and therapeutic intervention. METHODS: We conducted a prospective, observational study in adult patients (n = 72) sustaining severe injury admitted to a Level I Trauma Center. Healthy volunteers (n = 13) were also examined. Fecal specimens were collected on admission to the emergency department and at 3, 7, 10, and 13 days (±2 days) following injury. Microbial DNA was isolated for 16s rRNA sequencing, and α and ß diversities were estimated, according to taxonomic classification against the Greengenes database. RESULTS: The gut microbiome of trauma patients was altered on admission (i.e., within 30 minutes following injury) compared to healthy volunteers. Patients with an unchanged gut microbiome on admission were transfused more RBCs than those with an altered gut microbiome (p < 0.001). Although the gut microbiome started to return to a ß-diversity profile similar to that of healthy volunteers over time, it remained different from healthy controls. Alternatively, α diversity initially increased postinjury, but subsequently decreased during the hospitalization. Injured patients on admission had a decreased abundance of traditionally beneficial microbial phyla (e.g., Firmicutes) with a concomitant decrease in opportunistic phyla (e.g., Proteobacteria) compared to healthy controls (p < 0.05). Large amounts of blood products and RBCs were both associated with higher α diversity (p < 0.001) and a ß diversity clustering closer to healthy controls. CONCLUSION: The human gut microbiome changes early after trauma and may be aided by early massive transfusion. Ultimately, the gut microbiome of trauma patients may provide valuable diagnostic and therapeutic insight for the improvement of outcomes postinjury. LEVEL OF EVIDENCE: Prognostic and Epidemiological, level III.


Subject(s)
Blood Volume/physiology , Erythrocyte Transfusion , Gastrointestinal Microbiome/physiology , Wounds, Nonpenetrating/physiopathology , Wounds, Nonpenetrating/therapy , Wounds, Penetrating/physiopathology , Wounds, Penetrating/therapy , Adult , Bacterial Load , Correlation of Data , Feces/microbiology , Female , Humans , Injury Severity Score , Intestinal Mucosa/physiopathology , Male , Middle Aged , Prognosis , Prospective Studies , Wounds, Nonpenetrating/diagnosis
7.
Shock ; 52(2): 240-248, 2019 08.
Article in English | MEDLINE | ID: mdl-29953417

ABSTRACT

The microbiome is defined as the collective genomes of the microbes (composed of bacteria, bacteriophage, fungi, protozoa, and viruses) that colonize the human body, and alterations have been associated with a number of disease states. Changes in gut commensals can influence the neurologic system via the brain-gut axis, and systemic insults such as trauma or traumatic brain injury (TBI) may alter the gut microbiome. The objective of this study was to evaluate the gut microbiome in a preclinical TBI cortical impact model. Male rats underwent craniotomy and randomized to a sham group (n = 4), or a moderate TBI (n = 10) using a pneumatic impactor. MRI and behavioral assessments were performed pre-TBI and on days 2, 7, and 14 days thereafter. Microbiome composition was determined with 16s rRNA sequencing from fecal sample DNA pre-TBI and 2 hrs, 1, 3, and 7 days afterward. Alpha- and ß-bacterial diversity, as well as organizational taxonomic units (OTUs), were determined. Significant changes in the gut microbiome were evident as early as 2 h after TBI as compared with pre-injured samples and sham rats. While there were varying trends among the phylogenetic families across time, some changes persisted through 7 days in the absence of therapeutic intervention. While large structural lesions and behavioral deficits were apparent post-TBI, there were modest but significant decreases in α-diversity. Moreover, both changes in representative phyla and α-diversity measures were significantly correlated with MRI-determined lesion volume. These results suggest that changes in the microbiome may represent a novel biomarker to stage TBI severity and predict functional outcome.


Subject(s)
Brain Injuries, Traumatic/microbiology , Gastrointestinal Microbiome/physiology , Analysis of Variance , Animals , Gastrointestinal Microbiome/genetics , Male , Principal Component Analysis , RNA, Ribosomal, 16S/genetics , Rats , Time Factors
8.
Am J Surg ; 216(4): 699-705, 2018 10.
Article in English | MEDLINE | ID: mdl-30100050

ABSTRACT

BACKGROUND: This study characterizes the gastrointestinal (GI) microbiome in a pre-clinical polytrauma hemorrhage model. METHODS: Rats (n = 6) were anesthetized, hemorrhaged 20% of their blood volume, and subjected to a femur fracture and crush injuries to the small intestine, liver, and limb skeletal muscle without resuscitation. Fecal samples were collected pre-injury and 2 h post-injury. Purified DNA from the samples underwent 16s rRNA sequencing for microbial quantification. Bacterial diversity analysis and taxonomic classification were performed. RESULTS: Following injury, the gut microbial composition was altered with a shift in beta diversity and significant differences in the relative abundance of taxa. The relative abundance of the families Lachnospiraceae and Mogibacteriaceae was increased at 2 h, while Barnesiellaceae and Bacteroidaceae were decreased. Alpha diversity was unchanged. CONCLUSIONS: The GI microbiome is altered in rats subjected to a polytrauma hemorrhage model at 2 h post-injury in the absence of antibiotics or therapeutic interventions.


Subject(s)
Gastrointestinal Microbiome , Hemorrhage/microbiology , Multiple Trauma/microbiology , Animals , Hemorrhage/etiology , Multiple Trauma/complications , Rats , Rats, Sprague-Dawley
9.
Blood Coagul Fibrinolysis ; 29(1): 48-54, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28957941

ABSTRACT

: Alterations in coagulation, inflammation and immunity are associated with major injury. As platelets have both coagulation and immune functions, the aim of this study is to correlate platelet activation with the immunoinflammatory response in trauma and burn patients. Blood samples were drawn from trauma and burn patients and healthy volunteers. Platelet (sCD40L) and coagulation (D-dimers) activation, cytokines and inflammatory markers were assessed. sCD40L, D-dimers and cytokines were elevated in both injury groups. Overall, sCD40L levels correlated with interleukin (IL)-6 and tumour necrosis factor-alpha. Subanalysis revealed a correlation between sCD40L and IL-17a in the healthy volunteers and burn groups, but not the trauma group. A parallel activation of platelets and the inflammatory response occurs postinjury. However, in trauma patients, a potential critical interrelationship between platelet activation and the Th-17 response appears to be lacking, which may contribute to coagulopathic and immunoinflammatory complications and warrants further study.


Subject(s)
Inflammation/immunology , Platelet Activation/immunology , Th17 Cells/immunology , Wounds and Injuries/immunology , Cytokines/blood , Female , Humans , Male , Prospective Studies
10.
PLoS One ; 12(6): e0179015, 2017.
Article in English | MEDLINE | ID: mdl-28575063

ABSTRACT

BACKGROUND: Burn-induced inflammation leads to impaired immune responses resulting in increased morbidity and mortality. T-cells are central in the immune response and circulating CD4 and CD8 T-cells have been used to evaluate immune status; however, the role of these T-cell subsets in the burn wound is unknown. METHODS: Male C57BL/6 mice were subjected to a major 3rd degree scald burn or sham treatment. Twenty-four hours later, full thickness skin samples from sham mice and the burn wounds were collected and single cells were isolated and analyzed for αß TCR, γδ TCR, CD3, CD4, CD8 and CD69 expressions by flow cytometry. RESULTS: The burn wound contained significantly greater numbers of T-cells than skin from sham mice, due to a profound infiltration of αß T-cells. These infiltrating αß T-cells were primarily suppressor T-cells with a CD8+ or CD8-CD4- phenotype. The 15-fold increase in CD8+ αß T-cells caused a decrease in the CD4:CD8 ratio from 0.7 in sham skin to 0.3 in the burn wound. In contrast, the majority of the γδ T-cells in sham skin were CD4-CD8-, which decreased 9-fold in the burn wound. CD69 expression was suppressed on burn wound αß T-cells, but increased on γδ T-cells in the burn wound. CONCLUSIONS: The infiltrating burn wound αß T-cells likely act to quell inflammation. In contrast wound γδ T-cells were activated with elevated CD4 and CD69 expression. Thus, these two distinct T-cell subsets likely differentially regulate the burn wound inflammatory response.


Subject(s)
Burns/pathology , Skin/pathology , T-Lymphocyte Subsets/pathology , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/immunology , Burns/immunology , CD3 Complex/analysis , CD3 Complex/immunology , CD4 Antigens/analysis , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8 Antigens/analysis , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Lectins, C-Type/analysis , Lectins, C-Type/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/analysis , Receptors, Antigen, T-Cell, gamma-delta/immunology , Skin/immunology , T-Lymphocyte Subsets/immunology
11.
Burns ; 43(2): 297-303, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28341255

ABSTRACT

Burns are associated with activation of the innate immunity that can contribute to complications. Damage-associated molecular patterns (DAMPs) released after tissue injury play a critical role in the activation of the innate immunity, which appears to be mediated via toll-like receptors (TLRs). Previous findings have shown that TLRs and TLR-mediated responses are up-regulated after burn. Nonetheless, it is unclear what impact burn has on circulating levels of DAMPs. To study this, male C57BL/6 mice were subjected to a major burn or sham procedure. Three hours to 7days thereafter, plasma was collected and assayed for the representative DAMPs (i.e., HMGB1, cytochrome C, DNA and S100A) and extracellular cleavage products (fibronectin and hyaluronan). HMGB1, cytochrome C, fibronectin and hyaluronan levels were elevated in a time-dependent manner after burn as compared to sham levels. A significant elevation in TNF-α, IL-6 and IL-10 cytokine plasma levels was also found after burn. All cytokine levels were increased as early as 3h and remained elevated up to 24h. Circulating CD11b+ monocytes were increased at 24h after burn and showed increased expression of TLR-2. In conclusion, these findings support the concept that burn-induced elevations in circulating DAMPs are in part responsible for monocyte activation and the development of inflammatory complications under such conditions and warrants further investigation.


Subject(s)
Alarmins/blood , Burns/metabolism , Inflammation/metabolism , Monocytes/metabolism , Animals , Biomarkers/blood , Cytochromes c/blood , Disease Models, Animal , Fibronectins/blood , HMGB1 Protein/blood , Hyaluronic Acid/blood , Interleukin-10/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/blood
12.
Shock ; 47(6): 680-687, 2017 06.
Article in English | MEDLINE | ID: mdl-27941590

ABSTRACT

OBJECTIVES: Autotransfusion of shed blood from traumatic hemothorax is an attractive option for resuscitation of trauma patients in austere environments. However, previous analyses revealed that shed hemothorax (HX) blood is defibrinated, thrombocytopenic, and contains elevated levels of D-dimer. Mixing studies with normal pooled plasma demonstrated hypercoagulability, evoking concern for potentiation of acute traumatic coagulopathy. We hypothesized that induction of coagulopathic changes by shed HX blood may be due to increases in cellular microparticles (MP) and that these may also affect recipient platelet function. METHODS: Shed HX blood was obtained from 17 adult trauma patients under an Institutional Review Board approved prospective observational protocol. Blood samples were collected every hour up to 4 h after thoracostomy tube placement. The corresponding plasma was isolated and frozen for analysis. The effects of shed HX frozen plasma (HFP) and isolated HX microparticles (HMP) on coagulation and platelet function were assessed through mixing studies with platelet-rich plasma at various dilutions followed by analysis with thromboelastometry (ROTEM), platelet aggregometry (Multiplate), enzyme-linked immunosorbent assays, and flow cytometry. Furthermore, HFP was assessed for von Willebrand factor antigen levels and multimer content, and plasma-free hemoglobin. RESULTS: ROTEM analysis demonstrated that diluted HFP and isolated HMP samples decreased clotting time, clotting formation time, and increased α angle, irrespective of sample concentrations, when compared with diluted control plasma. Isolated HMP inhibited platelet aggregation in response to adenosine diphosphate, arachidonic acid, and collagen. HFP contained elevated levels of fibrin-degradation products and tissue factor compared with control fresh frozen plasma samples. MP concentrations in HFP were significantly increased and enriched in events positive for phosphatidylserine, tissue factor, CD235, CD45, CD41a, and CD14. von Willebrand factor (vWF) multimer analysis revealed significant loss of high molecular weight multimers in HFP samples. Plasma-free hemoglobin levels were 8-fold higher in HFP compared with fresh frozen plasma. CONCLUSION: HFP induces plasma hypercoagulability that is likely related to increased tissue factor and phosphatidylserine expression originating from cell-derived MP. In contrast, platelet dysfunction is induced by HMP, potentially aggravated by depletion of high molecular weight multimers of vWF. Thus, autologous transfusion of shed traumatic hemothorax blood may induce a range of undesirable effects in patients with acute traumatic coagulopathy.


Subject(s)
Cell-Derived Microparticles/metabolism , Hemothorax/metabolism , Platelet Aggregation/physiology , Wounds and Injuries/metabolism , Adult , Blood Coagulation Factors/metabolism , Female , Flow Cytometry , Humans , Immunoassay , Male , Middle Aged , Platelet Function Tests , Prospective Studies , Thrombelastography , Young Adult
13.
Shock ; 47(4): 500-505, 2017 04.
Article in English | MEDLINE | ID: mdl-27648695

ABSTRACT

INTRODUCTION: Severe trauma, hemorrhage, and resuscitation can lead to a trauma-related acute lung injury that involves rapid infiltration of immune cells and platelets. This infiltration involves exymatic degradation of matrix proteins, including plasmin, and causes loss of barrier function. Since tranexamic acid (TXA) inhibits plasminogen/ plasmin binding to target substrates, it may attenuate loss of barrier function after severe trauma, hemorrhage, and resuscitation. METHODS: Sprague-Dawley rats were subjected to polytrauma (laparotomy, and trauma to intestines, liver, right leg skeletal muscle, and right femur fracture), then bled 40% of their blood volume. One hour after completion of polytrauma and hemorrhage, resuscitation was begun with fresh whole blood (FWB) or FWB with prior bolus administration of TXA (10 mg/kg in 0.2 mL). RESULTS: Polytrauma, hemorrhage, and resuscitation with FWB led to an elevation in lung water content that was significantly reduced with TXA administration. Polytrauma and hemorrhage led to rise in the number of neutrophils/monocytes and platelets in the lungs, and a rise in myeloperoxidase (MPO), neutrophil elastase and complement C5a content. While resuscitation with FWB significantly reduced the cellular infiltrate and MPO, FWB/TXA further reduced the levels of neutrophil/monocytes, neutrophil elastase, and complement C5a. Polytrauma and hemorrhage led to rise in lung plasmin activity that was significantly reduced with either FWB or FWB/TXA resuscitation. CONCLUSION: Severe trauma and hemorrhage leads to increases in lung water content, and immune cell, platelets, MPO, elastase, and C5a content in lung tissue, all markers of inflammation and acute lung injury. The addition of TXA to FWB resuscitation markedly attenuated the rise in these parameters suggesting its utility in treating acute lung injury.


Subject(s)
Hemorrhage/drug therapy , Multiple Trauma/drug therapy , Tranexamic Acid/therapeutic use , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Hemorrhage/metabolism , Lung/drug effects , Lung/metabolism , Male , Multiple Trauma/metabolism , Rats , Rats, Sprague-Dawley , Resuscitation/methods
14.
J Surg Res ; 206(1): 139-145, 2016 11.
Article in English | MEDLINE | ID: mdl-27916353

ABSTRACT

BACKGROUND: Inflammation and activation of the innate immune system are often associated with traumatic injury and may involve alterations in toll-like receptor (TLR)-mediated responses. METHODS: A prospective observational study was designed and conducted. Twenty-one severely injured (ISS = 16-41) trauma intensive care unit (ICU) patients and six healthy volunteers that served as controls were enrolled. Anticoagulated whole blood was collected at 2-12 d after ICU admission and incubated in the presence of media alone (baseline), zymosan (TLR2 agonist) or lipopolysaccharide (LPS; TLR4 agonist) for 3 h. Supernatant levels of inflammatory cytokines (IL-1ß, IL-6, IL-10, and TNFα) were determined. RESULTS: TLR2-mediated and TLR4-mediated activation of whole blood cell cultures from both healthy volunteers and subjects-induced elevated cytokine levels over that observed in unstimulated cultures. Baseline values of IL-6 were significantly elevated in subject cultures as compared to healthy volunteers. Healthy volunteer cultures had 2-3-fold greater levels of IL-6 and TNFα than subject cultures when stimulated with zymosan (TLR2 agonist) or LPS (TLR4 agonist). IL-1ß and IL-10 levels did not differ significantly between healthy volunteers and subjects. CONCLUSIONS: The ability of circulating leukocytes from trauma ICU patients to be activated by TLR agonists is markedly suppressed and may play a role in the development of subsequent infectious complications.


Subject(s)
Cytokines/blood , Leukocytes/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Wounds and Injuries/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Case-Control Studies , Female , Humans , Intensive Care Units , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Male , Middle Aged , Prospective Studies , Salmonella , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Wounds and Injuries/blood , Young Adult , Zymosan/pharmacology
15.
PLoS One ; 11(7): e0158993, 2016.
Article in English | MEDLINE | ID: mdl-27403524

ABSTRACT

BACKGROUND: Gamma delta T-cells have been shown to be important to the early immunoinflammatory response to injury, independent of infection. This unique T-cell population acts to regulate cell trafficking and the release of cytokines and growth factors. We propose this sterile inflammatory response is in part associated with damage associated molecular patterns (DAMPs) generated by major injury, such as burn, and mediated via toll-like receptors (TLRs). It is unknown whether DAMPs can activate resident γδ T-cells that reside in skin. METHODS: Gamma delta T-cells were isolated from the skin of male C57BL/6 mice by enzymatic digestion. Mitochondrial DAMPs (MTDs) were generated from mitochondria isolated from mouse livers by sonication and centrifugation. Dermal γδ T-cells were incubated with MTDs (0-500 µg/ml) for 24 hr and cells and supernatants were collected for analysis. RESULTS: MTDs activated dermal γδ T-cells, as evidenced by increased TLR2 and TLR4 expression following in vitro exposure. MTDs also induced the production of inflammatory cytokines (IL-1ß, IL-6), and growth factors (PDGF and VEGF) by γδ T-cells. CONCLUSIONS: These findings herein support the concept that MTDs released after tissue/cellular injury are capable of activating dermal γδ T-cells. We propose that the activation of this unique T-cell population is central in the initiation of sterile inflammation and also contributes to the subsequent healing processes.


Subject(s)
Mitochondria/pathology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Skin/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Burns/immunology , Burns/pathology , Cytokines/biosynthesis , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/biosynthesis , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptors/metabolism
16.
Shock ; 46(3 Suppl 1): 108-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27172150

ABSTRACT

BACKGROUND: Acute lung injury (ALI) has been observed clinically after severe trauma. We have recently developed a rat model of polytrauma that shows evidence of multi-organ failure and coagulopathy. In this study, we investigate whether ALI occurs after severe trauma and resuscitation, and the cellular mechanisms involved. METHODS: Polytrauma and hemorrhage was induced in anesthetized Sprague-Dawley rats. Five groups were prepared: control, no resuscitation, and resuscitation with Lactated Ringer (LR), fresh whole blood or whole blood stored 7days at 4°C. Resuscitation was begun 1 hr after trauma. Lung injury was determined by lung wet/dry weight ratios. RESULTS: Polytrauma and hemorrhage (no resuscitation) led to a significant increase in the number of neutrophils, monocytes, macrophages, platelets, and the levels of myeloperoxidase, pro-inflammatory cytokines (IL-6, IL-1α, IL-1ß), anti-inflammatory Th2 cytokines (IL-4, IL-10, IL-13), and chemokines (MIP-1α, GRO KC) in the lung tissue. Resuscitation with LR, fresh whole blood or stored blood led to a significant change in the lung wet/dry ratio signifying fluid movement into the lungs. However, fluid did not move into the lungs in non-resuscitated controls. CONCLUSION: This study shows that trauma related acute lung injury occurs early after polytrauma and hemorrhage in rat. This ALI is secondary to the trauma, and likely due to an elevation in leukocytes, platelets, inflammatory cytokines and myeloperoxidase in the lung tissue prior to any resuscitation. Resuscitation with either LR or whole blood demonstrated similar lung edema. Blood was neither more protective nor more damaging than LR during early resuscitation.


Subject(s)
Acute Lung Injury/pathology , Acute Lung Injury/therapy , Wounds and Injuries/complications , Acute Lung Injury/etiology , Animals , Blood Platelets/metabolism , Cytokines/metabolism , Edema/complications , Edema/metabolism , Edema/pathology , Fluorescent Antibody Technique , Hemorrhage/mortality , Hemorrhage/pathology , Hemorrhage/therapy , Leukocytes/metabolism , Lung/immunology , Lung/metabolism , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Resuscitation , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
17.
Shock ; 46(2): 144-8, 2016 08.
Article in English | MEDLINE | ID: mdl-26974427

ABSTRACT

PURPOSE: The autotransfusion of unwashed (or unprocessed) shed hemothorax blood (USHB) in trauma patients is widely assumed to be beneficial; however, the inflammatory potential of shed pleural blood has not been thoroughly studied. Since previous studies have documented marked changes in coagulation function of shed pleural blood, we hypothesized that its level of inflammatory cytokines would be elevated. METHODS: A prospective observational study of trauma patients in whom cytokine levels from USHB were compared to venous samples from healthy volunteers was conducted. Differences between the cytokine content of patient-derived samples were compared to those from healthy subjects. RESULTS: There was a statistically significant increase in pro-inflammatory cytokines (IL-6, IL-8, TNFα, GM-CSF), a pro-inflammatory Th-1 cytokine (IFNγ), and anti-inflammatory Th-2 cytokines (IL-4 and IL-10) in shed pleural blood over four hours when compared with samples from healthy controls (P <0.05). Cytokine levels in USHB are approximately 10- to 100-fold higher compared with healthy control venous samples. CONCLUSIONS: USHB, even collected within the accepted four-hour window, contains significantly elevated cytokine levels, suggesting the potential for deleterious effects from autotransfusion. Randomized trials are needed to determine the safety and efficacy of autotransfusion in trauma patients.


Subject(s)
Cytokines/blood , Hemothorax/blood , Thoracic Injuries/blood , Thoracic Injuries/immunology , Wounds and Injuries/blood , Wounds and Injuries/immunology , Adult , Blood Transfusion, Autologous/adverse effects , Female , Humans , Interleukin-10/blood , Interleukin-4/blood , Interleukin-6/blood , Interleukin-8/blood , Male , Middle Aged , Prospective Studies , Tumor Necrosis Factor-alpha/blood
18.
Cytokine ; 76(2): 328-333, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26364992

ABSTRACT

BACKGROUND: The overall immunopathology of the T-helper cell (Th)-17 immune response has been implicated in various inflammatory diseases including pulmonary inflammation; however its potential role in acute respiratory distress syndrome (ARDS) is not defined. This study aimed to evaluate the Th-17 response in bronchoalveolar lavage fluid (BALF) and blood and from trauma patients with pulmonary complications. METHODS: A total of 21 severely injured intensive care unit (ICU) subjects, who were mechanically ventilated and undergoing bronchoscopy, were enrolled. BALF and blood were collected and analyzed for Th-1 (interferon [IFN]γ), Th-2 (interleukin [IL]-4, -10), Th-17 (IL-17A, -17F, -22, 23) and pro-inflammatory (IL-1ß, IL-6, tumor necrosis factor [TNF]α) cytokine levels. RESULTS: Significant levels of the Th-17 cytokines IL-17A, -17F and -21 and IL-6 (which can be classified as a Th-17 cytokine) were observed in the BALF of all subjects. There were no significant differences in Th-17 cytokines between those subjects with ARDS and those without, with the exception of plasma and BALF IL-6, which was markedly greater in ARDS subjects, as compared with controls and non-ARDS subjects. CONCLUSIONS: Trauma patients with pulmonary complications exhibited a significant Th-17 response in the lung and blood, suggesting that this pro-inflammatory milieu may be a contributing factor to such complications.


Subject(s)
Intensive Care Units , Respiratory Distress Syndrome/complications , Th17 Cells/immunology , Wounds and Injuries/complications , Adult , Bronchoalveolar Lavage Fluid , Case-Control Studies , Cytokines/blood , Cytokines/metabolism , Female , Humans , Male , Middle Aged , Prospective Studies , Respiratory Distress Syndrome/immunology , Wounds and Injuries/immunology , Young Adult
19.
Int J Burns Trauma ; 5(2): 56-65, 2015.
Article in English | MEDLINE | ID: mdl-26309777

ABSTRACT

Trauma-related pain is a natural consequence of injury and its surgical management; however, the relationship between opiates and complications in trauma patients is unknown. To study this a retrospective chart review of selected subjects following traumatic injury with admission to the SICU for > 3 days was performed, and opiate administration data was collected for the first 3 days of admission. Associated data from each subject's chart was also collected. Analysis of the data revealed that increased opiate intake after admission to the SICU was associated with significantly increased SICU and hospital LOS independent of injury severity. This increase in LOS was independent of mechanical ventilation in the moderate ISS group. Infectious complications were also more prevalent in the moderate ISS group with higher opiate use. These findings suggest that increased doses of opiate analgesics in trauma patients may contribute to an increased overall LOS and associated infectious complications. Analgesic regimes that minimize opiate intake, while still providing adequate pain relief, may be advantageous in reducing LOS, complications and reduce hospitalization costs.

20.
Shock ; 44 Suppl 1: 129-37, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25664984

ABSTRACT

Severe trauma can lead to a coagulopathy in patients, which is associated with increased mortality. We developed a rat polytrauma model that demonstrates a similar progression of coagulopathy. Because coagulation is influenced by changes in inflammation, and this interrelationship is poorly understood, we have studied the progression of inflammation, and its correlation with coagulation, in this rat model of severe polytrauma. Sprague-Dawley rats were anesthetized with isoflurane. Polytrauma was induced by damaging 10 cm of small intestines, right and medial liver lobes, right leg skeletal muscle, femur fracture, and hemorrhaging 40% of blood volume. No resuscitation was given. Polytrauma and hemorrhage resulted in a significant decrease in the number of lymphocytes and an increase in monocytes and granulocytes. There was an increase in plasma proinflammatory cytokines: tumor necrosis factor α (40×), interleukin (IL)-6 (20×), IL-1ß (16×), IL-17 (15×), interferon γ (10×), IL-1α (8×) and IL-12p70 (5×); anti-inflammatory cytokines: IL-10 (100×), IL-13 (16×), and IL-4 (5×); chemokines: growth-regulated protein/keratinocyte chemoattractant (30×), macrophage inflammatory protein 3α (10×), regulated and normal T-cell expressed and secreted (3×); and growth factors: vascular endothelial growth factor (5×), granulocyte macrophage colony-stimulating factor (6×), macrophage colony-stimulating factor (3×), granulocyte colony-stimulating factor (2×), and IL-5 (3×). There was a strong and significant correlation between prothrombin time, activated partial thromboplastin time, fibrinogen, and fibrin monomer concentration, and many cytokines. Polytrauma with hemorrhage is associated with a coagulopathy and a complex inflammatory response consisting of a concurrent rise in both proinflammatory and anti-inflammatory cytokines. The rise in plasma concentrations of chemokines and growth factors likely contribute to the mobilization of monocytes and granulocytes. There is strong correlation between prothrombin time, activated partial thromboplastin time, and IL-10 and IL-1ß. This relationship could be exploited for the development of resuscitation strategies that attenuate these cytokines and allow for better outcomes in patients with trauma through concomitant modulation of inflammation and coagulopathy.


Subject(s)
Blood Coagulation Disorders/therapy , Inflammation/blood , Animals , Blood Coagulation , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Granulocytes/cytology , Hemodynamics , Inflammation/metabolism , Lymphocytes/cytology , Male , Monocytes/cytology , Multiple Trauma/blood , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...