Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Front Immunol ; 14: 1227751, 2023.
Article En | MEDLINE | ID: mdl-37520569

Acute respiratory distress syndrome (ARDS) is a leading cause of morbidity and mortality in polytrauma patients. Pharmacological treatments of ARDS are lacking, and ARDS patients rely on supportive care. Accurate diagnosis of ARDS is vital for early intervention and improved outcomes but is presently delayed up to days. The use of biomarkers for early identification of ARDS development is a potential solution. Inflammatory mediators high-mobility group box 1 (HMGB1), syndecan-1 (SDC-1), and C3a have been previously proposed as potential biomarkers. For this study, we analyzed these biomarkers in animals undergoing smoke inhalation and 40% total body surface area burns, followed by intensive care for 72 h post-injury (PI) to determine their association with ARDS and mortality. We found that the levels of inflammatory mediators in serum were affected, as well as the degree of HMGB1 and Toll-like receptor 4 (TLR4) signal activation in the lung. The results showed significantly increased HMGB1 expression levels in animals that developed ARDS compared with those that did not. Receiver operating characteristic (ROC) analysis showed that HMGB1 levels at 6 h PI were significantly associated with ARDS development (AUROC=0.77) and mortality (AUROC=0.82). Logistic regression analysis revealed that levels of HMGB1 ≥24.10 ng/ml are associated with a 13-fold higher incidence of ARDS [OR:13.57 (2.76-104.3)], whereas the levels of HMGB1 ≥31.39 ng/ml are associated with a 12-fold increase in mortality [OR: 12.00 (2.36-93.47)]. In addition, we found that mesenchymal stem cell (MSC) therapeutic treatment led to a significant decrease in systemic HMGB1 elevation but failed to block SDC-1 and C3a increases. Immunohistochemistry analyses showed that smoke inhalation and burn injury induced the expression of HMGB1 and TLR4 and stimulated co-localization of HMGB1 and TLR4 in the lung. Interestingly, MSC treatment reduced the presence of HMGB1, TLR4, and the HMGB1-TLR4 co-localization. These results show that serum HMGB1 is a prognostic biomarker for predicting the incidence of ARDS and mortality in swine with smoke inhalation and burn injury. Therapeutically blocking HMGB1 signal activation might be an effective approach for attenuating ARDS development in combat casualties or civilian patients.


Burns , HMGB1 Protein , Respiratory Distress Syndrome , Smoke Inhalation Injury , Swine , Animals , Toll-Like Receptor 4 , Prognosis , HMGB1 Protein/metabolism , Respiratory Distress Syndrome/therapy , Smoke Inhalation Injury/complications , Smoke Inhalation Injury/therapy , Burns/complications , Biomarkers , Smoke
2.
Int J Mol Sci ; 24(8)2023 Apr 19.
Article En | MEDLINE | ID: mdl-37108656

Trauma-related hemorrhagic shock (HS) remains a leading cause of death among military and civilian trauma patients. We have previously shown that administration of complement and HMGB1 inhibitors attenuate morbidity and mortality 24 h after injury in a rat model of blast injury (BI) and HS. To further validate these results, this study aimed to develop a swine model and evaluate BI+HS-induced pathophysiology. Anesthetized Yucatan minipigs underwent combined BI and volume-controlled hemorrhage. After 30 min of shock, animals received an intravenous bolus of PlasmaLyte A and a continuous PlasmaLyte A infusion. The survival rate was 80% (4/5), and the non-survivor expired 72 min post-BI. Circulating organ-functional biomarkers, inflammatory biomarkers, histopathological evaluation, and CT scans indicated evidence of multiple-organ damage, systemic innate immunological activation, and local tissue inflammation in the injured animals. Interestingly, a rapid and dramatic increase in plasma levels of HMGB1 and C3a and markedly early myocarditis and encephalitis were associated with early death post-BI+HS. This study suggests that this model reflects the immunopathological alterations of polytrauma in humans during shock and prolonged damage control resuscitation. This experimental protocol could be helpful in the assessment of immunological damage control resuscitation approaches during the prolonged care of warfighters.


Blast Injuries , HMGB1 Protein , Shock, Hemorrhagic , Humans , Rats , Animals , Swine , Disease Models, Animal , Swine, Miniature , Hemorrhage , Shock, Hemorrhagic/therapy
3.
Front Immunol ; 14: 1100461, 2023.
Article En | MEDLINE | ID: mdl-37006238

The hallmark of acute respiratory distress syndrome (ARDS) pathobiology is unchecked inflammation-driven diffuse alveolar damage and alveolar-capillary barrier dysfunction. Currently, therapeutic interventions for ARDS remain largely limited to pulmonary-supportive strategies, and there is an unmet demand for pharmacologic therapies targeting the underlying pathology of ARDS in patients suffering from the illness. The complement cascade (ComC) plays an integral role in the regulation of both innate and adaptive immune responses. ComC activation can prime an overzealous cytokine storm and tissue/organ damage. The ARDS and acute lung injury (ALI) have an established relationship with early maladaptive ComC activation. In this review, we have collected evidence from the current studies linking ALI/ARDS with ComC dysregulation, focusing on elucidating the new emerging roles of the extracellular (canonical) and intracellular (non-canonical or complosome), ComC (complementome) in ALI/ARDS pathobiology, and highlighting complementome as a vital nexus of the pathobiological connectome for ALI/ARDS via its crosstalking with other systems of the immunome, DAMPome, PAMPome, coagulome, metabolome, and microbiome. We have also discussed the diagnostic/therapeutic potential and future direction of ALI/ARDS care with the ultimate goal of better defining mechanistic subtypes (endotypes and theratypes) through new methodologies in order to facilitate a more precise and effective complement-targeted therapy for treating these comorbidities. This information leads to support for a therapeutic anti-inflammatory strategy by targeting the ComC, where the arsenal of clinical-stage complement-specific drugs is available, especially for patients with ALI/ARDS due to COVID-19.


Acute Lung Injury , COVID-19 , Connectome , Respiratory Distress Syndrome , Humans , Lung/pathology , Respiratory Distress Syndrome/drug therapy , Acute Lung Injury/pathology , Complement System Proteins/therapeutic use
4.
Br J Pharmacol ; 180(4): 422-440, 2023 02.
Article En | MEDLINE | ID: mdl-36251578

BACKGROUND AND PURPOSE: Traumatic haemorrhage (TH) is the leading cause of potentially preventable deaths that occur during the prehospital phase of care. No effective pharmacological therapeutics are available for critical TH patients yet. Here, we identify terminal complement activation (TCA) as a therapeutic target in combat casualties and evaluate the efficacy of a TCA inhibitor (nomacopan) on organ damage and survival in vivo. EXPERIMENTAL APPROACH: Complement activation products and cytokines were analysed in plasma from 54 combat casualties. The correlations between activated complement pathway(s) and the clinical outcomes in trauma patients were assessed. Nomacopan was administered to rats subjected to lethal TH (blast injury and haemorrhagic shock). Effects of nomacopan on TH were determined using survival rate, organ damage, physiological parameters, and laboratory profiles. KEY RESULTS: Early TCA was associated with systemic inflammatory responses and clinical outcomes in this trauma cohort. Lethal TH in the untreated rats induced early TCA that correlated with the severity of tissue damage and mortality. The addition of nomacopan to a damage-control resuscitation (DCR) protocol significantly inhibited TCA, decreased local and systemic inflammatory responses, improved haemodynamics and metabolism, attenuated tissue and organ damage, and increased survival. CONCLUSION AND IMPLICATIONS: Previous findings of our and other groups revealed that early TCA represents a rational therapeutic target for trauma patients. Nomacopan as a pro-survival and organ-protective drug, could emerge as a promising adjunct to DCR that may significantly reduce the morbidity and mortality in severe TH patients while awaiting transport to critical care facilities.


Complement C5 , Shock, Hemorrhagic , Rats , Animals , Complement C5/pharmacology , Shock, Hemorrhagic/drug therapy , Complement Activation , Immunologic Factors/pharmacology , Phenotype
5.
Front Immunol ; 13: 991048, 2022.
Article En | MEDLINE | ID: mdl-36341368

Complementopathy, endotheliopathy, and coagulopathy following a traumatic injury are key pathophysiological mechanisms potentially associated with multiple-organ failure (MOF) and mortality. However, the heterogeneity in the responses of complementopathy, endotheliopathy, and coagulopathy to trauma, the nature and extent of their interplay, and their relationship to clinical outcomes remain unclear. Fifty-four poly-trauma patients were enrolled and divided into three subgroups based on their ISS. Biomarkers in blood plasma reflecting complement activation, endothelial damage, and coagulopathy were measured starting from admission to the emergency department and at 3, 6, 12, 24, and 120 hours after admission. Comparative analyses showed that severely injured patients (ISS>24) were associated with longer days on mechanical ventilation, in the intensive care unit and hospital stays, and a higher incidence of hyperglycemia, bacteremia, respiratory failure and pneumonia compared to mildly (ISS<16) or moderately (ISS=16-24) injured patients. In this trauma cohort, complement was activated early, primarily through the alternative complement pathway. As measured in blood plasma, severely injured patients had significantly higher levels of complement activation products (C3a, C5a, C5b-9, and Bb), endothelial damage markers (syndecan-1, sTM, sVEGFr1, and hcDNA), and fibrinolytic markers (D-dimer and LY30) compared to less severely injured patients. Severely injured patients also had significantly lower thrombin generation (ETP and peak) and lower levels of coagulation factors (I, V, VIII, IX, protein C) than less severely injured patients. Complement activation correlated with endothelial damage and hypocoagulopathy. Logistic regression analyses revealed that Bb >1.57 µg/ml, syndecan-1 >66.6 ng/ml or D-dimer >6 mg/L at admission were associated with a higher risk of MOF/mortality. After adjusting for ISS, each increase of the triadic score defined above (Bb>1.57 µg/ml/Syndecan-1>66.6 ng/ml/D-dimer>6.0mg/L) was associated with a 6-fold higher in the odds ratio of MOF/death [OR: 6.83 (1.04-44.96, P=0.046], and a 4-fold greater in the odds of infectious complications [OR: 4.12 (1.04-16.36), P=0.044]. These findings provide preliminary evidence of two human injury response endotypes (traumatized triad and non-traumatized triad) that align with clinical trajectory, suggesting a potential endotype defined by a high triadic score. Patients with this endotype may be considered for timely intervention to create a pro-survival/organ-protective phenotype and improve clinical outcomes.


Blood Coagulation Disorders , Multiple Trauma , Humans , Syndecan-1 , Injury Severity Score , Prospective Studies , Blood Coagulation Disorders/etiology , Biomarkers , Multiple Trauma/complications , Multiple Organ Failure
6.
Trauma Surg Acute Care Open ; 7(1): e000927, 2022.
Article En | MEDLINE | ID: mdl-36117727

Objectives: Early complementopathy and coagulopathy are shown often after trauma. However, the prevalence of any interplay between complement cascade (ComC) and coagulation cascade (CoaC) after trauma remains unclear. This study intended to explore whether complement-coagulation crosstalk exists, which may provide a reliable guide to clinical implications in trauma patients. Methods: This single-center cohort study of trauma patients enrolled 100 patients along with 20 healthy volunteers. Blood samples from patients were collected at admission, 45, 90, 135 minutes, and 18 hours after admission. Demographic characteristics were recorded, blood levels of ComC and CoaC factors, and inflammatory cytokines were measured by ELISA, clot-based assays, or luminex multiplex assay, and partial thromboplastin (PT) and partial thromboplastin time (PTT) were assessed using a Behring blood coagulation system. Results: Compared with the healthy controls, plasma levels of complement factors (C5b-9 and Bb) and 11 tested inflammatory cytokines increased in moderately and severely injured patients as early as 45 minutes after admission and sustained higher levels up to 18 hours after admission. C5b-9 correlated positively to patients' hospital stay. In parallel, the consumption of coagulation factors I, II, X, and XIII was shown throughout the first 18 hours after admission in moderately and severely injured patients, whereas PT, PTT, D-dimer, factor VII, and factor VIII values significantly increased from the admission to 135 minutes in moderately and severely injured patients. Along with an inverse correlation between plasma Bb, factors I and II, a positive correlation between C5b-9, Bb, D-dimer, PT, and PTT was evident. Conclusions: This study demonstrates trauma-induced early activation of plasma cascades including ComC, CoaC, and fibrinolytic cascade, and their correlation between plasma cascades in severe trauma patients. Our study suggests that the simultaneous modulation of plasma cascades might benefit clinical outcomes for trauma patients. Level of evidence: Prospective study, level III.

7.
Biomolecules ; 12(1)2022 01 08.
Article En | MEDLINE | ID: mdl-35053249

Several preclinical and clinical reports have demonstrated that levels of circulating high mobility group box 1 protein (HMGB1) are increased early after trauma and are associated with systemic inflammation and clinical outcomes. However, the mechanisms of the interaction between HMGB1 and inflammatory mediators that lead to the development of remote organ damage after trauma remain obscure. HMGB1 and inflammatory mediators were analyzed in plasma from 54 combat casualties, collected on admission to a military hospital in Iraq, and at 8 and 24 h after admission. In total, 45 (83%) of these patients had traumatic brain injury (TBI). Nine healthy volunteers were enrolled as controls. HMGB1 plasma levels were significantly increased in the first 8 h after admission, and were found to be associated with systemic inflammatory responses, injury severity score, and presence of TBI. These data provided the rationale for designing experiments in rats subjected to blast injury and hemorrhage, to explore the effect of HMGB1 inhibition by CX-01 (2-O, 3-O desulfated heparin). Animals were cannulated, then recovered for 5-7 days before blast injury in a shock tube and volume-controlled hemorrhage. Blast injury and hemorrhage induced an early increase in HMGB1 plasma levels along with severe tissue damage and high mortality. CX-01 inhibited systemic HMGB1 activity, decreased local and systemic inflammatory responses, significantly reduced tissue and organ damage, and tended to increase survival. These data suggest that CX-01 has potential as an adjuvant treatment for traumatic hemorrhage.


Brain Injuries, Traumatic , HMGB1 Protein , Animals , Brain Injuries, Traumatic/metabolism , HMGB1 Protein/metabolism , Humans , Inflammation/metabolism , Rats
8.
PLoS One ; 15(11): e0242450, 2020.
Article En | MEDLINE | ID: mdl-33201908

BACKGROUND AND OBJECTIVE: Resuscitative Endovascular Balloon Occlusion of Aorta (REBOA) has emerged as a potential life-saving maneuver for the management of non-compressible torso hemorrhage in trauma patients. Complete REBOA (cREBOA) is inherently associated with the burden of ischemia reperfusion injury (IRI) and organ dysfunction. However, the distal organ inflammation and its association with organ injury have been little investigated. This study was conducted to assess these adverse effects of cREBOA following massive hemorrhage in swine. METHODS: Spontaneously breathing and consciously sedated Sinclair pigs were subjected to exponential hemorrhage of 65% total blood volume over 60 minutes. Animals were randomized into 3 groups (n = 7): (1) Positive control (PC) received immediate transfusion of shed blood after hemorrhage, (2) 30min-cREBOA (A30) received Zone 1 cREBOA for 30 minutes, and (3) 60min-cREBOA (A60) given Zone 1 cREBOA for 60 minutes. The A30 and A60 groups were followed by resuscitation with shed blood post-cREBOA and observed for 4h. Metabolic and hemodynamic effects, coagulation parameters, inflammatory and end organ consequences were monitored and assessed. RESULTS: Compared with 30min-cREBOA, 60min-cREBOA resulted in (1) increased IL-6, TNF-α, and IL-1ß in distal organs (kidney, jejunum, and liver) (p < 0.05) and decreased reduced glutathione in kidney and liver (p < 0.05), (2) leukopenia, neutropenia, and coagulopathy (p < 0.05), (3) blood pressure decline (p < 0.05), (4) metabolic acidosis and hyperkalemia (p < 0.05), and (5) histological injury of kidney and jejunum (p < 0.05) as well as higher levels of creatinine, AST, and ALT (p < 0.05). CONCLUSION: 30min-cREBOA seems to be a feasible and effective adjunct in supporting central perfusion during severe hemorrhage. However, prolonged cREBOA (60min) adverse effects such as distal organ inflammation and injury must be taken into serious consideration.


Balloon Occlusion/adverse effects , Resuscitation/methods , Shock, Hemorrhagic/physiopathology , Animals , Aorta/physiopathology , Balloon Occlusion/methods , Blood Pressure , Blood Pressure Determination , Blood Transfusion , Endovascular Procedures/adverse effects , Endovascular Procedures/methods , Hemodynamics , Hemorrhage , Inflammation , Liver/physiopathology , Male , Models, Animal , Reperfusion Injury/physiopathology , Shock, Hemorrhagic/metabolism , Swine , Torso/physiopathology
9.
Mil Med ; 184(Suppl 1): 265-272, 2019 03 01.
Article En | MEDLINE | ID: mdl-30901455

OBJECTIVE: Mild blast traumatic brain injury is commonly prevalent in modern combat casualty care and has been associated with the development of neurodegenerative conditions. However, whether primary lower level blast overpressure (LBOP) causes neurodegeneration and neuroinflammation remains largely unknown. The aim of our present study was to determine whether LBOP can cause neuroinflammation and neurodegeneration. METHODS: Anesthetized rats were randomly assigned to LBOP group (70 kPa, n = 5) or sham group (without blast, n = 5). Histopathological and cytokine changes in brain tissue at 5 days post-injury were evaluated by hematoxylin-eosin staining and Bioplex assay, respectively. RESULTS: Histopathological assessment revealed neuronal degeneration and increased density of inflammatory cells in frontal and parietal cortex, hippocampus and thalamus in rats exposed to LBOP. LBOP exposure significantly elevated levels of pro-inflammatory cytokines (EPO, IL-1ß, IL-6, IL-12, IL-18, and TNF-α) and chemokines (GRO and RANTES) as well as of an anti-inflammatory cytokine (IL-13) in the frontal cortex. CONCLUSIONS: This study reveals a role of neuroinflammation in neurodegeneration after mild blast traumatic brain injury. Therapies that target this process might in warfighters might function either by attenuating the development of post-traumatic stress disorder, chronic traumatic encephalopathy and Alzheimer's disease, or by slowing their progression.


Encephalitis/pathology , Explosions/statistics & numerical data , Nerve Degeneration/pathology , Animals , Biomarkers/analysis , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Chemokine CCL5/analysis , Chemokine CXCL1/analysis , Chemokines/analysis , Cytokines/analysis , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Interleukin-12/analysis , Interleukin-18/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Rats/injuries , Tumor Necrosis Factor-alpha/analysis
10.
Mil Med ; 184(Suppl 1): 282-290, 2019 03 01.
Article En | MEDLINE | ID: mdl-30901474

OBJECTIVE: Blast injury is associated with multi-organ failure (MOF), causing significant morbidity and mortality in trauma patients. However, the pathogenesis of blast-induced MOF still remains obscure. In this study, we evaluate the pathophysiological changes related to blast-induced MOF in a clinically relevant rat model of blast injury. METHODS: A moderate blast overpressure was applied to induce injury in anesthetized rats. Pathological changes were evaluated by H&E staining. Complement activation, plasminogen, and myeloperoxidase levels were analyzed by complement hemolytic assay (CH50) and/or ELISA in blood samples. RESULTS: Analysis of lung, brain, and liver tissue at 24 hour after blast overpressure revealed severe injuries. The level of complement components C3 and C1q decreased in parallel with the reduction of CH50 level in injured animals at 1, 3, and 6 hours after blast. Consumption of plasminogen was also detected as early as 1 hour post-injury. Myeloperoxidase levels were elevated within 1 hour of blast injury. CONCLUSION: Our data reveal that blast injury triggers the complement and fibrinolytic systems, which likely contribute to blast-induced MOF. Conceivably, therapies that target these systems early may improve clinical outcomes in blast patients.


Blast Injuries/drug therapy , Fibrinolytic Agents/pharmacokinetics , Multiple Organ Failure/etiology , Animals , Blast Injuries/complications , Brain/drug effects , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Fibrinolytic Agents/blood , Fibrinolytic Agents/therapeutic use , Injury Severity Score , Liver/drug effects , Liver/pathology , Liver/physiopathology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Multiple Organ Failure/drug therapy , Rats , Rats, Sprague-Dawley/blood , Statistics, Nonparametric
11.
PLoS One ; 13(8): e0202594, 2018.
Article En | MEDLINE | ID: mdl-30133517

BACKGROUND AND OBJECTIVE: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action. METHODS: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500µg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR. RESULTS: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs. CONCLUSIONS: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.


Acute Lung Injury/drug therapy , Blast Injuries/drug therapy , CD55 Antigens/administration & dosage , HMGB1 Protein/genetics , Inflammation/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/physiopathology , Animals , Blast Injuries/genetics , Blast Injuries/pathology , Complement Activation/drug effects , Complement C3a/antagonists & inhibitors , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/physiopathology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , NF-kappa B/genetics , Pressure/adverse effects , Rats , Rats, Sprague-Dawley
12.
Indian J Med Microbiol ; 35(4): 522-528, 2017.
Article En | MEDLINE | ID: mdl-29405144

PURPOSE: The aim of this study was to predict the T-cell and B-cell epitopes in major outer membrane protein (MOMP) of Chlamydia trachomatis (CT) by using online software and also to analyse the secondary structure of MOMP through bioinformatics tools. MATERIALS AND METHODS: The predictions of secondary structure of MOMP protein were carried out using SOPMA software, and the prediction of B-cell epitopes in MOMP protein was carried out using IEDB and LEPS software, while the T-cell epitopes were predicted by the software of IEBD and SYFPEITHI. The predictions from the software were combined with MOMP protein characteristics, including surface features, hydrophilicity, flexibility, accessibility and plasticity, to analyse the common epitope areas' response by T-cells and B-cells. RESULTS: In the secondary structure of CT MOMP, the alpha-helices accounted for 41.62% of total amino acid, while the beta sheets and random coil accounted for 19.80% and 32.49%, respectively. Predictions combined with MOMP protein surface features, hydrophilicity, flexibility, accessibility and plasticity were further characterised, and three high-score B-cell epitope areas were found as located in 24-31, 307-311 and 318-327 amino acids of MOMP protein, respectively; in the meanwhile, three high-score T-cell epitope areas were found in 234-236, 323-329 and 338-343 amino acids of MOMP using major histocompatibility complex (MHC) class I HLA-A 0201 restrictive T-cell epitope analyser. CONCLUSION: We established the methods by using the biological information network technologies for looking the T-cell antigen epitopes and B-cell antigen epitopes in MOMP of CT, and three novel T-cell epitopes as well as three novel B-cell epitopes were identified in the current study. It provides important information for further studying the antigenicity of CT MOMP protein and also provides useful information for developing highly efficient subunit vaccines for CT.


Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Computational Biology , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/genetics , Bacterial Outer Membrane Proteins/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Models, Molecular , Protein Conformation , Software
13.
Microbes Infect ; 18(11): 669-674, 2016 Nov.
Article En | MEDLINE | ID: mdl-27436813

Ascending infection by sexually transmitted Chlamydia trachomatis is required for chlamydial induction of tubal pathology. To achieve ascension, the C. trachomatis organisms may have to spread from cell to cell, which inevitably exposes the organisms to extracellular mucosal effectors such as complement factors that are known to possess strong antichlamydial activities. Here, we report that the chlamydia-secreted protease CPAF efficiently neutralized complement factor C3-dependent antichlamydial activity. The neutralization was dependent on the proteolytic activity of CPAF and correlated with the CPAF-mediated degradation of complement factor C3 and factor B. As a result, CPAF preferentially inhibited the alternative complement activation pathway. The significance and limitation of these observations were discussed.


Chlamydia trachomatis/immunology , Complement C3/antagonists & inhibitors , Complement Factor B/antagonists & inhibitors , Endopeptidases/immunology , Immune Evasion , Immunologic Factors/antagonists & inhibitors , Complement Pathway, Alternative , HeLa Cells , Humans , Proteolysis
14.
Infect Immun ; 84(9): 2697-702, 2016 09.
Article En | MEDLINE | ID: mdl-27382018

Despite the extensive in vitro characterization of CPAF (chlamydial protease/proteasome-like activity factor), its role in chlamydial infection and pathogenesis remains unclear. We now report that a Chlamydia trachomatis strain deficient in expression of CPAF (L2-17) is no longer able to establish a successful infection in the mouse lower genital tract following an intravaginal inoculation. The L2-17 organisms were cleared from the mouse lower genital tract within a few days, while a CPAF-sufficient C. trachomatis strain (L2-5) survived in the lower genital tract for more than 3 weeks. However, both the L2-17 and L2-5 organisms maintained robust infection courses that lasted up to 4 weeks when they were directly delivered into the mouse upper genital tract. The CPAF-dependent chlamydial survival in the lower genital tract was confirmed in multiple strains of mice. Thus, we have demonstrated a critical role of CPAF in promoting C. trachomatis survival in the mouse lower genital tracts. It will be interesting to further investigate the mechanisms of the CPAF-dependent chlamydial pathogenicity.


Chlamydia Infections/microbiology , Chlamydia trachomatis/metabolism , Endopeptidases/metabolism , Genitalia/microbiology , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Line, Tumor , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL
15.
Infect Immun ; 84(2): 467-79, 2016 02.
Article En | MEDLINE | ID: mdl-26597987

We previously associated a missense mutation of the tc0668 gene of serial in vitro-passaged Chlamydia muridarum, a murine model of human urogenital C. trachomatis, with severely attenuated disease development in the upper genital tract of female mice. Since these mutants also contained a TC0237 Q117E missense mutation that enhances their in vitro infectivity, an effort was made here to isolate and characterize a tc0668 single mutant to determine its individual contribution to urogenital pathogenicity. Detailed genetic analysis of C. muridarum passages revealed a truncated variant with a G216* nonsense mutation of the 408-amino-acid TC0668 protein that does not produce a detectable product. Intracellular growth and infectivity of C. muridarum in vitro remain unaffected in the absence of TC0668. Intravaginal inoculation of the TC0668 null mutant into C3H/HeJ mice results in a typical course of lower genital tract infection but, unlike a pathogenic isogenic control, is unable to elicit significant chronic inflammation of the oviduct and fails to induce hydrosalpinx. Thus, TC0668 is demonstrated as an important chromosome-encoded urogenital pathogenicity factor of C. muridarum and the first with these characteristics to be discovered for a Chlamydia pathogen.


Chlamydia muridarum/genetics , Chlamydia muridarum/pathogenicity , Reproductive Tract Infections/microbiology , Virulence Factors/genetics , Animals , Chlamydia Infections/microbiology , Chlamydia muridarum/growth & development , Codon, Nonsense , DNA, Bacterial/genetics , Disease Models, Animal , Fallopian Tubes/immunology , Fallopian Tubes/microbiology , Female , Humans , Mice, Inbred C3H , Molecular Sequence Data , Phenotype , Reproductive Tract Infections/pathology
16.
Pathog Dis ; 74(2)2016 Mar.
Article En | MEDLINE | ID: mdl-26676260

Chlamydia trachomatis causes sexually transmitted diseases with infertility, pelvic inflammatory disease and neonatal pneumonia as complications. The duration of urogenital mouse models with the strict mouse pathogen C. muridarum addressing vaginal shedding, pathological changes of the upper genital tract or infertility is rather long. Moreover, vaginal C. trachomatis application usually does not lead to the complications feared in women. A fast-to-perform mouse model is urgently needed to analyze new antibiotics, vaccine candidates, immune responses (in gene knockout animals) or mutants of C. trachomatis. To complement the valuable urogenital model with a much faster and quantifiable screening method, we established an optimized lung infection model for the human intracellular bacterium C. trachomatis serovar D (and L2) in immunocompetent C57BL/6J mice. We demonstrated its usefulness by sensitive determination of antibiotic effects characterizing advantages and limitations achievable by early or delayed short tetracycline treatment and single-dose azithromycin application. Moreover, we achieved partial acquired protection in reinfection with serovar D indicating usability for vaccine studies, and showed a different course of disease in absence of complement factor C3. Sensitive monitoring parameters were survival rate, body weight, clinical score, bacterial load, histological score, the granulocyte marker myeloperoxidase, IFN-γ, TNF-α, MCP-1 and IL-6.


Anti-Bacterial Agents/therapeutic use , Bacterial Vaccines/immunology , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/physiology , Chlamydial Pneumonia/drug therapy , Chlamydial Pneumonia/prevention & control , Host-Pathogen Interactions , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Load , Biopsy , Cell Line , Chlamydial Pneumonia/microbiology , Chlamydial Pneumonia/mortality , Complement C3/genetics , Complement C3/immunology , Cytokines/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin G/immunology , Lung/drug effects , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Knockout , Peroxidase/metabolism
17.
Infect Immun ; 83(12): 4701-9, 2015 Dec.
Article En | MEDLINE | ID: mdl-26416907

Chlamydia trachomatis infection in the lower genital tract can ascend to and cause pathologies in the upper genital tract, potentially leading to severe complications, such as tubal infertility. However, chlamydial organisms depleted of plasmid or deficient in the plasmid-encoded Pgp3 are attenuated in ascending infection and no longer are able to induce the upper genital tract pathologies, indicating a significant role of Pgp3 in chlamydial pathogenesis. We now report that C. trachomatis Pgp3 can neutralize the antichlamydial activity of human cathelicidin LL-37, a host antimicrobial peptide secreted by both genital tract epithelial cells and infiltrating neutrophils. Pgp3 bound to and formed stable complexes with LL-37. We further showed that the middle region of Pgp3 (Pgp3m) was responsible for both the binding to and neutralization of LL-37, suggesting that Pgp3m can be targeted for attenuating chlamydial pathogenicity or developed for blocking LL-37-involved non-genital-tract pathologies, such as rosacea and psoriasis. Thus, the current study has provided significant information for both understanding the mechanisms of chlamydial pathogenesis and developing novel therapeutic agents.


Antigens, Bacterial/immunology , Antimicrobial Cationic Peptides/immunology , Bacterial Proteins/immunology , Chlamydia trachomatis/immunology , Plasmids/immunology , Virulence Factors/immunology , Amino Acid Sequence , Antigens, Bacterial/genetics , Antimicrobial Cationic Peptides/genetics , Bacterial Proteins/genetics , Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Host-Pathogen Interactions , Humans , Molecular Sequence Data , Mutation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Virulence Factors/genetics , Cathelicidins
18.
Infect Immun ; 83(9): 3568-77, 2015 Sep.
Article En | MEDLINE | ID: mdl-26099591

Intravaginal infection with Chlamydia muridarum in mice can ascend to the upper genital tract, resulting in hydrosalpinx, a pathological hallmark for tubal infertility in women infected with C. trachomatis. Here, we utilized in vivo imaging of C. muridarum infection in mice following an intravaginal inoculation and confirmed the rapid ascent of the chlamydial organisms from the lower to upper genital tracts. Unexpectedly, the C. muridarum-derived signal was still detectable in the abdominal area 100 days after inoculation. Ex vivo imaging of the mouse organs revealed that the long-lasting presence of the chlamydial signal was restricted to the gastrointestinal (GI) tract, which was validated by directly measuring the chlamydial live organisms and genomes in the same organs. The C. muridarum organisms spreading from the genital to the GI tracts were detected in different mouse strains and appeared to be independent of oral or rectal routes. Mice prevented from orally taking up excretions also developed the long-lasting GI tract infection. Inoculation of C. muridarum directly into the upper genital tract, which resulted in a delayed vaginal shedding of live organisms, accelerated the chlamydial spreading to the GI tract. Thus, we have demonstrated that the genital tract chlamydial organisms may use a systemic route to spread to and establish a long-lasting infection in the GI tract. The significance of the chlamydial spreading from the genital to GI tracts is discussed.


Chlamydia Infections/pathology , Gastrointestinal Tract/microbiology , Animals , Chlamydia muridarum , Female , Fluorescent Antibody Technique , Mice , Real-Time Polymerase Chain Reaction
19.
PLoS One ; 10(4): e0124840, 2015.
Article En | MEDLINE | ID: mdl-25915629

We have previously shown that the plasmid-encoded Pgp3 is a major virulence factor for C. muridarum induction of hydrosalpinx. We now report that Pgp5 also plays a significant role in the development of hydrosalpinx following C. muridarum induction. Pgp5 deficiency was introduced via either in-frame deletion (CM-Δpgp5) or premature stop codon installation (CM-pgp5S). Mice infected with either CM-Δpgp5 or CM-pgp5S developed hydrosalpinges at significantly reduced levels with an incidence rate of <40% and a mean severity score of 2 or less. In contrast, 80% or more mice developed hydrosalpinx with a severity score of >3 when mice were infected with Pgp5-sufficient C. muridarum (plasmid-competent wild type or plasmid-free C. muridarum transformed with a full plasmid or depleted of pgp7 gene). The attenuated pathogenicity of the Pgp5-deficient C. muridarum correlated with a significantly reduced level of ascending infection in the oviduct tissue despite the similar overall shedding courses between mice infected with Pgp5-deficient versus sufficient C. muridarum. Furthermore, in the oviducts of mice infected with Pgp5-deficient C. muridarum, significantly lower levels of inflammatory cell infiltration and cytokine production were detected. Thus, Pgp5 is a significant plasmid-encoded virulence factor for C. muridarum pathogenicity in the upper genital tract.


Chlamydia Infections/microbiology , Chlamydia muridarum/pathogenicity , Fallopian Tubes/microbiology , Glycoproteins/metabolism , Virulence Factors/metabolism , Animals , Chlamydia muridarum/genetics , Female , Glycoproteins/genetics , Mice , Plasmids/genetics , Virulence Factors/genetics
20.
Infect Immun ; 83(6): 2327-37, 2015 Jun.
Article En | MEDLINE | ID: mdl-25824829

Although Chlamydia-induced hydrosalpinx in women and mice has been used as a surrogate marker for tubal infertility, the medical relevance of nontubal pathologies, such as uterine horn dilation, developed in mice following chlamydial infection remains unclear. We now report that the uterine horn dilation correlates with glandular duct dilation detected microscopically following Chlamydia muridarum infection. The dilated glandular ducts pushed the uterine horn lumen to closure or dilation and even broke through the myometrium to develop extrusion outside the uterine horn. The severity scores of uterine horn dilation observed macroscopically correlated well with the number of cross sections of the dilated glandular ducts counted under microscopy. Chlamydial infection was detected in the glandular epithelial cells, potentially leading to inflammation and dilation of the glandular ducts. Direct delivery of C. muridarum into the mouse uterus increased both uterine horn/glandular duct dilation and hydrosalpinx. However, the chlamydial plasmid, which is essential for the induction of hydrosalpinx, was not required for the induction of uterine horn/glandular duct dilation. Screening 12 strains of mice for uterine horn dilation following C. muridarum infection revealed that B10.D2, C57BL/10J, and C57BL/6J mice were most susceptible, followed by BALB/cJ and A/J mice. Deficiency in host genes involved in immune responses failed to significantly alter the C. muridarum induction of uterine horn dilation. Nevertheless, the chlamydial induction of uterine horn/glandular duct dilation may be used to evaluate plasmid-independent pathogenicity of Chlamydia in susceptible mice.


Chlamydia Infections/microbiology , Chlamydia muridarum , Epithelial Cells/microbiology , Uterine Diseases/microbiology , Uterus/pathology , Animals , Chlamydia Infections/pathology , Female , Gene Expression Regulation/physiology , Mice , Mice, Inbred Strains , Mice, Knockout , Uterine Diseases/pathology
...