Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.508
Filter
1.
BMC Pharmacol Toxicol ; 25(1): 41, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997770

ABSTRACT

BACKGROUND AND AIM: The use of cefoperazone/sulbactam (CPZ/SAM) could commonly cause vitamin K-dependent coagulation disorders and even hemorrhage sometimes. However, there is a lack of prediction tools estimating the risk for this. This study aimed at developing and internally validating a model for predicting CPZ/SAM-associated coagulation disorders in Chinese inpatients. METHODS: A case-control study was conducted in 11,092 adult inpatients admitted to a Chinese general hospital between 2020 and 2021 and treated with CPZ/SAM. Patients with CPZ/SAM-associated coagulation disorders were identified through the Adverse Drug Events Active Surveillance and Assessment System-II and subsequent manual evaluation. Controls were selected from eligible patients who didn't develop coagulation disorders after CPZ/SAM therapy, with a 1:1 propensity score matching. The final predictors were obtained by univariable and multivariable logistic regression analyses. Internal validation and calibration for the model were performed using 1000 bootstrap resamplings. RESULTS: 258 patients were identified as CPZ/SAM-associated coagulation disorders in 2184 patients eligible for inclusions and exclusions and the incidence was 11.8%. A final population of 252 cases and 252 controls was included for model development and validation. Malnutrition (OR = 2.41 (1.56-3.77)), history of recent bleeding (OR = 1.95 (1.32-2.90)), treatment duration (OR = 1.10 (1.07-1.14)), combination with carbapenems (OR = 4.43 (1.85-11.88)), and serum creatinine (OR = 1.01 (1.00-1.01)) were identified as final predictors. The model showed good discrimination, calibration, and clinical practicality, with the validated area under the receiver operating characteristic curve being 0.723 (0.683-0.770). CONCLUSIONS: The model with good performance quantifies the risk for CPZ/SAM-associated coagulation disorders, and may support individual assessment and interventions to mitigate the risk after external validation.


Subject(s)
Anti-Bacterial Agents , Blood Coagulation Disorders , Cefoperazone , Sulbactam , Humans , Cefoperazone/therapeutic use , Cefoperazone/adverse effects , Sulbactam/therapeutic use , Sulbactam/adverse effects , Female , Male , Middle Aged , Case-Control Studies , Aged , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , China , Blood Coagulation Disorders/chemically induced , Adult , Inpatients , East Asian People
3.
PLoS One ; 19(7): e0304231, 2024.
Article in English | MEDLINE | ID: mdl-38985805

ABSTRACT

Trauma is the leading cause of death in individuals up to 45 years of age. Alterations in platelet function are a critical component of trauma-induced coagulopathy (TIC), yet these changes and the potential resulting dysfunction is incompletely understood. The lack of clinical assays available to explore platelet function in this patient population has hindered detailed understanding of the role of platelets in TIC. The objective of this study was to assess trauma patient ex vivo flow-dependent platelet hemostatic capacity in a microfluidic model. We hypothesized that trauma patients would have flow-regime dependent alterations in platelet function. Blood was collected from trauma patients with level I activations (N = 34) within 60 min of hospital arrival, as well as healthy volunteer controls (N = 10). Samples were perfused through a microfluidic model of injury at venous and arterial shear rates, and a subset of experiments were performed after incubation with fluorescent anti-CD41 to quantify platelets. Complete blood counts were performed as well as plasma-based assays to quantify coagulation times, fibrinogen, and von Willebrand factor (VWF). Exploratory correlation analyses were employed to identify relationships with microfluidic hemostatic parameters. Trauma patients had increased microfluidic bleeding times compared to healthy controls. While trauma patient samples were able to deposit a substantial amount of clot in the model injury site, the platelet contribution to microfluidic hemostasis was attenuated. Trauma patients had largely normal hematology and plasma-based coagulation times, yet had elevated D-Dimer and VWF. Venous microfluidic bleeding time negatively correlated with VWF, D-Dimer, and mean platelet volume (MPV), while arterial microfluidic bleeding time positively correlated with oxygenation. Arterial clot growth rate negatively correlated with red cell count, and positively with mean corpuscular volume (MCV). We observed changes in clot composition in trauma patient samples reflected by significantly diminished platelet contribution, which resulted in reduced hemostatic function in a microfluidic model of vessel injury. We observed a reduction in platelet clot contribution under both venous and arterial flow ex vivo in trauma patient samples. While our population was heterogenous and had relatively mild injury severity, microfluidic hemostatic parameters correlated with different patient-specific data depending on the flow setting, indicating potentially differential mechanistic pathways contributing to platelet hemostatic capacity in the context of TIC. These data were generated with the goal of identifying key features of platelet dysfunction in bleeding trauma patients under conditions of flow and to determine if these features correlate with clinically available metrics, thus providing preliminary surrogate markers of physiological platelet dysfunction to be further studied across larger cohorts. Future studies will continue to explore those relationships and further define mechanisms of TIC and their relationship with patient outcomes.


Subject(s)
Blood Platelets , Hemostasis , Microfluidics , Wounds and Injuries , Humans , Blood Platelets/metabolism , Male , Female , Adult , Wounds and Injuries/blood , Wounds and Injuries/complications , Microfluidics/methods , Middle Aged , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/blood , von Willebrand Factor/metabolism , Fibrinogen/metabolism , Case-Control Studies , Bleeding Time
4.
Braz Oral Res ; 38: e052, 2024.
Article in English | MEDLINE | ID: mdl-38922212

ABSTRACT

The aim of this study was to evaluate the impact of oral conditions and health-related quality of life (HRQoL) on oral health-related quality of life (OHRQoL) in children and adolescents with blood coagulation disorders and hemoglobinopathies (BCDH). The study was cross-sectional and included 61 individuals aged 2 to 18 years with BCDH. Exams for dental caries (dmft/DMFT index), oral hygiene (simplified oral hygiene index - OHI-S), and gingival health (modified gingival index - MGI) were performed. The pediatric quality of life inventory™ (PedsQL™) generic core scale and oral health scale were used to measure HRQoL and OHRQoL. Spearman's correlation coefficient (ρ) and the Mann-Whitney test (α = 0.05) were conducted to assess the relationship between covariates and the PedsQL™ oral health scale. The mean PedsQL™ oral health scale score was 76.66 (SD = 21.36). Worse OHRQoL was correlated with poor oral hygiene (ρ = -0.383; p: 0.004), poor gingival health (ρ = -0.327; p = 0.014), and better HRQoL (ρ = 0.488; p < 0.001). Greater untreated dental caries experience was associated with worse OHRQoL (p = 0.009). Worse oral health status in children and adolescents with BCDH negatively impacts OHRQoL, and OHRQoL and quality of life analyzed from a generic perspective are positively correlated constructs in this population.


Subject(s)
Blood Coagulation Disorders , Dental Caries , Hemoglobinopathies , Oral Health , Quality of Life , Humans , Child , Adolescent , Female , Male , Cross-Sectional Studies , Oral Health/statistics & numerical data , Child, Preschool , Dental Caries/psychology , Hemoglobinopathies/psychology , Hemoglobinopathies/physiopathology , Hemoglobinopathies/complications , Blood Coagulation Disorders/psychology , Statistics, Nonparametric , Oral Hygiene Index , Periodontal Index , DMF Index , Surveys and Questionnaires , Socioeconomic Factors , Oral Hygiene
5.
Neurosurg Rev ; 47(1): 297, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922506

ABSTRACT

Coagulopathy development in traumatic brain injury (TBI) is among the significant complications that can negatively affect the clinical course and outcome of TBI patients. Timely identification of this complication is of utmost importance in the acute clinical setting. We reviewed TBI patients admitted to our trauma center from 2015 to 2021. Demographic data, mechanism of injury, findings on admission, imaging studies, procedures during hospitalization, and functional outcomes were gathered. INR with a cutoff of 1.3, platelet count less than 100 × 109/L, or partial thromboplastin time greater than 40s were utilized as the markers of coagulopathy. A total of 4002 patients were included. Coagulopathy occurred in 38.1% of the patients. Age of the patients (Odds Ratio (OR) = 0.993, 95% Confidence Interval (CI) = 0.986-0.999, p = 0.028), systolic blood pressure (OR = 0.993, 95% CI = 0.989-0.998, p = 0.005), fibrinogen level (OR = 0.998, 95% CI = 0.996-0.999, p < 0.001), and hemoglobin level (OR = 0.886, 95% CI = 0.839-0.936, p < 0.001) were independently associated with coagulopathy. Furthermore, coagulopathy was independently associated with higher mortality rates and longer ICU stays. Coagulopathy had the most substantial effect on mortality of TBI patients (OR = 2.6, 95% CI = 2.1-3.3, p < 0.001), compared to other admission clinical characteristics independently associated with mortality such as fixed pupillary light reflex (OR = 1.8, 95% CI = 1.5-2.4, p < 0.001), GCS (OR = 0.91, 95% CI = 0.88-0.94, p < 0.001), and hemoglobin level (OR = 0.93, 95% CI = 0.88-0.98, p = 0.004). Early coagulopathy in TBI patients can lead to higher mortality rates. Future studies are needed to prove that early detection and correction of coagulopathy and modifiable risk factors may help improve outcomes of TBI patients.


Subject(s)
Blood Coagulation Disorders , Brain Injuries, Traumatic , Humans , Brain Injuries, Traumatic/complications , Female , Male , Adult , Middle Aged , Retrospective Studies , Blood Coagulation Disorders/epidemiology , Blood Coagulation Disorders/etiology , Incidence , Aged , Risk Factors , Young Adult , Cohort Studies , Partial Thromboplastin Time
6.
J Infect Dev Ctries ; 18(5): 666-671, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38865388

ABSTRACT

INTRODUCTION: Coronavirus 2019 symptoms include coagulopathy and thromboembolic risk. Using one parameter to diagnose coagulopathy has little predictive value. OBJECTIVE: This study will examine if D-dimer and APTT testing can predict COVID-19 severity and aid triage and manage patients. METHODS: 214 COVID-19 patients were enrolled and classified into two categories based on their respiratory manifestations; mild (126 cases) and severe (88 cases). Patient data regarding age, gender, D-Dimer level, and APTT level were collected. When both D-Dimer and APTT levels were abnormal, in this study, the patient was considered to have a coagulation disorder. Indicators of coagulation in the COVID-19 patients were collected and compared between the two groups. Chi-square (χ2) tests were used to determine the significant differences between coagulation disorders in the two groups. RESULTS: Our findings showed that patients with coagulopathies were more likely to belong to the severe group. Within the two groups of patients, the rate of coagulation disorders was as follows: mild = 8.8 % within coagulation disorders, 4.8% within the two Groups; severe = 91.2 % within coagulation disorders, 77.8 % within the two Groups. There was a statistically significant relationship between coagulation disorder and severe COVID-19 patients compared to mild patients (p < 0.05). CONCLUSIONS: Coagulation disorders are more likely to occur in severe COVID-19 patients. D-Dimer and APTT tests are significant indicators for predicting COVID-19 severity. Our research found an abnormal pattern of coagulation disorders and COVID-19 severity that should be considered in the COVID-19 treatment protocol.


Subject(s)
Blood Coagulation Disorders , COVID-19 , Fibrin Fibrinogen Degradation Products , Predictive Value of Tests , Humans , COVID-19/blood , COVID-19/diagnosis , COVID-19/complications , Fibrin Fibrinogen Degradation Products/analysis , Male , Female , Middle Aged , Partial Thromboplastin Time , Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/blood , Adult , Aged , Severity of Illness Index , SARS-CoV-2/isolation & purification
7.
Shock ; 62(2): 265-274, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38888571

ABSTRACT

ABSTRACT: Background: Death due to hemorrhagic shock, particularly, noncompressible truncal hemorrhage, remains one of the leading causes of potentially preventable deaths. Automated partial and intermittent resuscitative endovascular balloon occlusion of the aorta (i.e., pREBOA and iREBOA, respectively) are lifesaving endovascular strategies aimed to achieve quick hemostatic control while mitigating distal ischemia. In iREBOA, the balloon is titrated from full occlusion to no occlusion intermittently, whereas in pREBOA, a partial occlusion is maintained. Therefore, these two interventions impose different hemodynamic conditions, which may impact coagulation and the endothelial glycocalyx layer. In this study, we aimed to characterize the clotting kinetics and coagulopathy associated with iREBOA and pREBOA, using thromboelastography (TEG). We hypothesized that iREBOA would be associated with a more hypercoagulopathic response compared with pREBOA due to more oscillatory flow. Methods: Yorkshire swine (n = 8/group) were subjected to an uncontrolled hemorrhage by liver transection, followed by 90 min of automated pREBOA, iREBOA, or no balloon support (control). Hemodynamic parameters were continuously recorded, and blood samples were serially collected during the experiment (i.e., eight key time points: baseline (BL), T0, T10, T30, T60, T90, T120, T210 min). Citrated kaolin heparinase assays were run on a TEG 5000 (Haemonetics, Niles, IL). General linear mixed models were employed to compare differences in TEG parameters between groups and over time using STATA (v17; College Station, TX), while adjusting for sex and weight. Results: As expected, iREBOA was associated with more oscillations in proximal pressure (and greater magnitudes of peak pressure) because of the intermittent periods of full aortic occlusion and complete balloon deflation, compared to pREBOA. Despite these differences in acute hemodynamics, there were no significant differences in any of the TEG parameters between the iREBOA and pREBOA groups. However, animals in both groups experienced a significant reduction in clotting times (R time: P < 0.001; K time: P < 0.001) and clot strength (MA: P = 0.01; G: P = 0.02) over the duration of the experiment. Conclusions: Despite observing acute differences in peak proximal pressures between the iREBOA and pREBOA groups, we did not observe any significant differences in TEG parameters between iREBOA and pREBOA. The changes in TEG profiles were significant over time, indicating that a severe hemorrhage followed by both pREBOA and iREBOA can result in faster clotting reaction times (i.e., R times). Nevertheless, when considering the significant reduction in transfusion requirements and more stable hemodynamic response in the pREBOA group, there may be some evidence favoring pREBOA usage over iREBOA.


Subject(s)
Balloon Occlusion , Disease Models, Animal , Resuscitation , Shock, Hemorrhagic , Thrombelastography , Animals , Swine , Balloon Occlusion/methods , Shock, Hemorrhagic/therapy , Resuscitation/methods , Blood Coagulation Disorders/therapy , Blood Coagulation Disorders/etiology , Blood Coagulation/drug effects , Hemorrhage/therapy , Hemodynamics , Female , Male
9.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(3): 852-856, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38926979

ABSTRACT

OBJECTIVE: To investigate the intervention effect and mechanism of regulating miR-155 on young rats with dysfunction of blood coagulation. METHODS: Twenty-six healthy and clean SD male rats were selected to establish the coagulopathy models. Twenty-four rats successfully established models and were randomly divided into three groups: model group, up-regulated miR-155 group and down-regulated miR-155 group, with 8 rats in each group. The expression of miR-155 was detected by real-time fluorescence quantitative polymerase chain reaction. The changes of coagulation factors and coagulation indicators were observed. Liver pathological tissues were observed by HE staining. The expressions of HMGB1-RAGE/TLRs-NF-κB signaling pathway related proteins were detected by Western blot. RESULTS: Compared with model group, the expressions of HMGB1, RAGE, TLR2, TLR4 and NF-κB were significantly increased in up-regulated miR-155 group (all P < 0.05), while decreased in down-regulated miR-155 group (all P < 0.05). Compared with model group, the expressions of coagulation factor Ⅱ, Ⅶ, Ⅸ, and Ⅹ were significantly decreased in up-regulated miR-155 group (all P < 0.05), while increased in down-regulated miR-155 group (P < 0.05). There was no significant difference in the expression of coagulation factor Ⅺ among the three groups (P >0.05). Compared with model group, the levels of prothrombin time (PT) and activated partial thromboplastin time (APTT) were lower and fibrinogen (FIB) was higher in up-regulated miR-155 group (all P < 0.05), while in the down-regulated miR-155 group they were opposite. CONCLUSION: Down-regulation of miR-155 can effectively improve coagulation factors and coagulation indexes and inhibit inflammation in young rats with dysfunction of blood coagulopathy, and the mechanism may be related to HMGB1-RAGE/TLRs-NF-κB signaling pathway.


Subject(s)
Blood Coagulation , HMGB1 Protein , MicroRNAs , NF-kappa B , Rats, Sprague-Dawley , Signal Transduction , Animals , Rats , Male , NF-kappa B/metabolism , HMGB1 Protein/metabolism , Blood Coagulation Disorders , Down-Regulation , Toll-Like Receptor 4/metabolism , Blood Coagulation Factors/metabolism , Toll-Like Receptor 2/metabolism
10.
Adv Gerontol ; 37(1-2): 149-152, 2024.
Article in Russian | MEDLINE | ID: mdl-38944786

ABSTRACT

In the treatment of coronavirus infections, it is important not only to understand the course of the disease, but also to understand what is happening in the human body, especially in the circulatory system, that is, which disorders lead to deterioration and further complications. Hemostasis disorder in COVID-19 plays an important role in the etiology and clinical manifestations of the disease. The ability to identify factors and risk groups for the development of thrombotic complications, the ability to dynamically interpret peripheral blood parameters and coagulograms, knowledge of diagnostic criteria for possible hemostasis disorders (for example, DIC syndrome, sepsis-associated coagulopathy, antiphospholipids, hemophagocytosis and hypercoagulation syndrome) are necessary to determine the indications for the test. Differentiated prescribing of clinically justified therapy (including anticoagulants and blood components) is important, which determines the complexity of treatment and prognosis for patients with COVID-19. This article is a review of the literature on the topic of hemostasis disorders in elderly and senile patients with mesenteric thrombosis in COVID 19 over the past few years.


Subject(s)
COVID-19 , SARS-CoV-2 , Thrombosis , Humans , COVID-19/complications , COVID-19/physiopathology , Aged , Thrombosis/etiology , Thrombosis/diagnosis , Thrombosis/blood , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/blood , Hemostasis/physiology , Anticoagulants/therapeutic use , Anticoagulants/administration & dosage
11.
PLoS Pathog ; 20(6): e1011777, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38913740

ABSTRACT

COVID-associated coagulopathy seemly plays a key role in post-acute sequelae of SARS- CoV-2 infection. However, the underlying pathophysiological mechanisms are poorly understood, largely due to the lack of suitable animal models that recapitulate key clinical and pathological symptoms. Here, we fully characterized AC70 line of human ACE2 transgenic (AC70 hACE2 Tg) mice for SARS-CoV-2 infection. We noted that this model is highly permissive to SARS-CoV-2 with values of 50% lethal dose and infectious dose as ~ 3 and ~ 0.5 TCID50 of SARS-CoV-2, respectively. Mice infected with 105 TCID50 of SARS-CoV-2 rapidly succumbed to infection with 100% mortality within 5 days. Lung and brain were the prime tissues harboring high viral titers, accompanied by histopathology. However, viral RNA and inflammatory mediators could be detectable in other organs, suggesting the nature of a systemic infection. Lethal challenge of AC70 hACE2 Tg mice caused acute onset of leukopenia, lymphopenia, along with an increased neutrophil-to-lymphocyte ratio (NLR). Importantly, infected animals recapitulated key features of COVID-19-associated coagulopathy. SARS-CoV-2 could induce the release of circulating neutrophil extracellular traps (NETs), along with activated platelet/endothelium marker. Immunohistochemical staining with anti-platelet factor-4 (PF4) antibody revealed profound platelet aggregates especially within blocked veins of the lungs. We showed that acute SARS-CoV-2 infection triggered a hypercoagulable state coexisting with ill-regulated fibrinolysis. Finally, we highlighted the potential role of Annexin A2 (ANXA2) in fibrinolytic failure. ANXA2 is a calcium-dependent phospholipid-binding protein that forms a heterotertrameric complexes localized at the extracellular membranes with two S100A10 small molecules acting as a co-receptor for tissue-plasminogen activator (t-PA), tightly involved in cell surface fibrinolysis. Thus, our results revealing elevated IgG type anti-ANXA2 antibody production, downregulated de novo ANXA2/S100A10 synthesis, and reduced ANXA2/S100A10 association in infected mice, this protein might serve as druggable targets for development of antithrombotic and/or anti-fibrinolytic agents to attenuate pathogenesis of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Disease Models, Animal , Mice, Transgenic , SARS-CoV-2 , Animals , COVID-19/pathology , COVID-19/complications , COVID-19/virology , COVID-19/metabolism , Mice , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Humans , Blood Coagulation Disorders/virology , Blood Coagulation Disorders/pathology , Pneumonia, Viral/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/metabolism , Betacoronavirus , Lung/virology , Lung/pathology , Lung/metabolism , Coronavirus Infections/virology , Coronavirus Infections/pathology , Coronavirus Infections/complications , Pandemics , Extracellular Traps/metabolism
12.
Article in English | MEDLINE | ID: mdl-38847555

ABSTRACT

This is an animal model study to investigate changes in hemostasis during endotoxemic shock and to determine whether the combination of inhaled nitric oxide (iNO) + intravenous hydrocortisone had an effect on clot formation and fibrinolysis. iNO selectively decreases pulmonary artery pressure, without affecting cardiac index or systemic vascular resistance; however, the results of studies on the possible consequences of iNO administration on coagulation are inconsistent and require further research. Thirty-four piglets were included. Administering endotoxin caused severe hypodynamic shock. Half of the animals received iNO (30 ppm) + hydrocortisone, starting 3 h after endotoxin infusion and continuing to the end of the study. All animals developed coagulation disorders, manifested by a tendency to hypocoagulation; at the same time, fibrinolysis was impaired. Coagulation and fibrinolysis disorders persisted after endotoxin infusion was discontinued, with worse severity in the animals that died before the study was terminated. Administering iNO + hydrocortisone did not cause further changes in coagulation and fibrinolysis parameters, either during or after the endotoxin challenge, suggesting that potential therapeutic interventions with iNO to lower pulmonary arterial pressure will not affect hemostasis.


Subject(s)
Blood Coagulation , Disease Models, Animal , Fibrinolysis , Hydrocortisone , Nitric Oxide , Shock, Septic , Thrombelastography , Animals , Hydrocortisone/administration & dosage , Hydrocortisone/therapeutic use , Hydrocortisone/pharmacology , Nitric Oxide/metabolism , Fibrinolysis/drug effects , Swine , Blood Coagulation/drug effects , Shock, Septic/drug therapy , Administration, Inhalation , Endotoxins/administration & dosage , Humans , Blood Coagulation Disorders/drug therapy
13.
J Innate Immun ; 16(1): 337-353, 2024.
Article in English | MEDLINE | ID: mdl-38815564

ABSTRACT

BACKGROUND: Sepsis-associated coagulopathy specifically refers to widespread systemic coagulation activation accompanied by a high risk of hemorrhage and organ damage, which in severe cases manifests as disseminated intravascular coagulation (DIC), or even develops into multiple organ dysfunction syndrome (MODS). The complement system and the coagulation system as the main columns of innate immunity and hemostasis, respectively, undergo substantial activation after sepsis. SUMMARY: Dysfunction of the complement, coagulation/fibrinolytic cascades caused by sepsis leads to "thromboinflammation," which ultimately amplifies the systemic inflammatory response and accelerates the development of MODS. Recent studies have revealed that massive activation of the complement system exacerbates sepsis-induced coagulation and even results in DIC, which suggests that inhibition of complement activation may have therapeutic potential in the treatment of septic coagulopathy. KEY MESSAGES: Sepsis-associated thrombosis involves the upregulation or activation of procoagulant factors, down-regulation or inactivation of anticoagulant factors, and impairment of the fibrinolytic mechanism. This review aims to summarize the latest literature and analyze the underlying molecular mechanisms of the activation of the complement system on the abnormal coagulation cascades in sepsis.


Subject(s)
Complement Activation , Sepsis , Humans , Sepsis/immunology , Complement Activation/immunology , Animals , Blood Coagulation , Disseminated Intravascular Coagulation/immunology , Disseminated Intravascular Coagulation/etiology , Immunity, Innate , Complement System Proteins/immunology , Complement System Proteins/metabolism , Multiple Organ Failure/immunology , Multiple Organ Failure/etiology , Fibrinolysis , Blood Coagulation Disorders/immunology , Blood Coagulation Disorders/etiology , Thrombosis/immunology , Thrombosis/etiology
14.
J Pak Med Assoc ; 74(5): 959-966, 2024 May.
Article in English | MEDLINE | ID: mdl-38783447

ABSTRACT

Sepsis is a potentially fatal illness marked by organ failure and the two main causes of which are shock and disseminated intravascular coagulation. Multi-organ dysfunction in sepsis is mediated by the inflammatory cytokine storm, while sepsis induced coagulopathy is mediated and accelerated by activation of pro-coagulative mechanisms. Regardless of the severity of sepsis, disseminated intravascular coagulation is a potent predictor of mortality in septic patients. Additionally, oxidative stress in sepsis causes renal ischaemia and eventually acute kidney injury. The first and foremost goal is to initiate resuscitation immediately, with treatment mainly focussing on maintaining a balance of coagulants and anticoagulants. A simpler and more universal diagnostic criteria is likely to improve studies on the spectrum associated with sepsis.


Subject(s)
Disseminated Intravascular Coagulation , Sepsis , Humans , Sepsis/complications , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/diagnosis , Disseminated Intravascular Coagulation/therapy , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/therapy , Multiple Organ Failure/etiology , Acute Kidney Injury/etiology , Acute Kidney Injury/therapy , Anticoagulants/therapeutic use , Oxidative Stress , Resuscitation/methods
15.
Clin Appl Thromb Hemost ; 30: 10760296241257517, 2024.
Article in English | MEDLINE | ID: mdl-38778544

ABSTRACT

Early identification of biomarkers that can predict the onset of sepsis-induced coagulopathy (SIC) in septic patients is clinically important. This study endeavors to examine the diagnostic and prognostic utility of serum C1q in the context of SIC. Clinical data from 279 patients diagnosed with sepsis at the Departments of Intensive Care, Respiratory Intensive Care, and Infectious Diseases at the Renmin Hospital of Wuhan University were gathered spanning from January 2022 to January 2024. These patients were categorized into two groups: the SIC group comprising 108 cases and the non-SIC group consisting of 171 cases, based on the presence of SIC. Within the SIC group, patients were further subdivided into a survival group (43 cases) and non-survival group (65 cases). The concentration of serum C1q in the SIC group was significantly lower than that in the non-SIC group. Furthermore, A significant correlation was observed between serum C1q levels and both SIC score and coagulation indices. C1q demonstrated superior diagnostic and prognostic performance for SIC patients, as indicated by a higher area under the curve (AUC). Notably, when combined with CRP, PCT, and SOFA score, C1q displayed the most robust diagnostic efficacy for SIC. Moreover, the combination of C1q with the SOFA score heightened predictive value concerning the 28-day mortality of SIC patients.


Subject(s)
Blood Coagulation Disorders , Complement C1q , Sepsis , Humans , Sepsis/blood , Sepsis/complications , Sepsis/diagnosis , Sepsis/mortality , Male , Female , Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/blood , Middle Aged , Complement C1q/metabolism , Prognosis , Aged , Biomarkers/blood
16.
J Spec Oper Med ; 24(2): 61-66, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38801744

ABSTRACT

BACKGROUND: The development of acute traumatic coagulopathy is associated with increased mortality and morbidity in patients with battlefield traumatic injuries. Currently, the incidence of acute traumatic coagulopathy in the Role 1 setting is unclear. METHODS: We queried the Prehospital Trauma Registry (PHTR) module of the Department of Defense Trauma Registry (DoDTR) for all encounters from inception through May 2019. The PHTR captures data on Role 1 prehospital care. Data from the PHTR was linked to the DoDTR to analyze laboratory data and patient outcomes using descriptive statistics. We defined coagulopathy as an international normalized ratio (INR) of ≥1.5 or platelet count ≤150×109/L. RESULTS: A total of 595 patients met the inclusion criteria; 36% (212) met our definition for coagulopathy, with 31% (185) carrying low platelet numbers, 11% (68) showing an elevated INR, and 7% (41) with both. The baseline (no coagulopathy) cohort had a mean INR of 1.10 (95% CI 1.09-1.12) versus 1.38 (95% CI 1.33-1.43) in the coagulopathic cohort. The mean platelet count was 218 (95% CI 213-223) ×109/L in the baseline cohort versus 117 (95% CI 110-125) ×109/L in the coagulopathic cohort. CONCLUSIONS: Our findings indicate a high incidence of coagulopathy in trauma patients. Approximately one-third of wounded patients had laboratory evidence of coagulopathy upon presentation to a forward medical care facility. Advanced diagnostic facilities are therefore needed to facilitate early diagnosis of acute traumatic coagulopathy. Blood products with a long shelf life can aid in early correction.


Subject(s)
Blood Coagulation Disorders , Emergency Medical Services , International Normalized Ratio , Registries , Resuscitation , Humans , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/epidemiology , Incidence , Male , Adult , Resuscitation/methods , Female , Emergency Medical Services/statistics & numerical data , Wounds and Injuries/complications , Wounds and Injuries/therapy , Wounds and Injuries/epidemiology , Platelet Count , Military Personnel/statistics & numerical data , United States/epidemiology
18.
BMC Urol ; 24(1): 113, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807082

ABSTRACT

BACKGROUND: Hemorrhage is a common complication of nephrostomy and percutaneous nephrolithotripsy, and it is caused by surgical factors. Here we report a rare case of hemorrhage caused by sepsis-related coagulation dysfunction. CASE PRESENTATION: A 72-years-old male patient with bilateral ureteral calculi accompanied by hydronephrosis and renal insufficiency developed sepsis and hemorrhage on the third day after bilateral nephrostomy. After vascular injury was excluded by DSA, the hemorrhage was considered to be sepsis-associated coagulopathy(SAC/SIC), finally the patient recovered well after active symptomatic treatment. CONCLUSIONS: In patients with sepsis and hemorrhage, SAC/SIC cannot be excluded even if coagulation function is slightly abnormal after surgical factors are excluded. For urologists who may encounter similar cases in their general urology practice, it is important to be aware of these unusual causes of hemorrhage.


Subject(s)
Blood Coagulation Disorders , Nephrostomy, Percutaneous , Sepsis , Humans , Male , Aged , Sepsis/etiology , Nephrostomy, Percutaneous/adverse effects , Blood Coagulation Disorders/etiology , Postoperative Hemorrhage/etiology
19.
Crit Care ; 28(1): 168, 2024 05 18.
Article in English | MEDLINE | ID: mdl-38762746

ABSTRACT

BACKGROUND: Critically injured patients need rapid and appropriate hemostatic treatment, which requires prompt identification of trauma-induced coagulopathy (TIC) upon hospital admission. We developed and validated the performance of a clinical score based on prehospital resuscitation parameters and vital signs at hospital admission for early diagnosis of TIC. METHODS: The score was derived from a level-1 trauma center registry (training set). It was then validated on data from two other level-1 trauma centers: first on a trauma registry (retrospective validation set), and then on a prospective cohort (prospective validation set). TIC was defined as a PTratio > 1.2 at hospital admission. Prehospital (vital signs and resuscitation care) and admission data (vital signs and laboratory parameters) were collected. We considered parameters independently associated with TIC in the score (binomial logistic regression). We estimated the score's performance for the prediction of TIC. RESULTS: A total of 3489 patients were included, and among these a TIC was observed in 22% (95% CI 21-24%) of cases. Five criteria were identified and included in the TIC Score: Glasgow coma scale < 9, Shock Index > 0.9, hemoglobin < 11 g.dL-1, prehospital fluid volume > 1000 ml, and prehospital use of norepinephrine (yes/no). The score, ranging from 0 and 9 points, had good performance for the identification of TIC (AUC: 0.82, 95% CI: 0.81-0.84) without differences between the three sets used. A score value < 2 had a negative predictive value of 93% and was selected to rule-out TIC. Conversely, a score value ≥ 6 had a positive predictive value of 92% and was selected to indicate TIC. CONCLUSION: The TIC Score is quick and easy to calculate and can accurately identify patients with TIC upon hospital admission.


Subject(s)
Blood Coagulation Disorders , Early Diagnosis , Wounds and Injuries , Humans , Female , Male , Adult , Middle Aged , Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/etiology , Cohort Studies , Prospective Studies , Wounds and Injuries/complications , Wounds and Injuries/blood , Retrospective Studies , Registries/statistics & numerical data , Aged , Hospitalization/statistics & numerical data
20.
Neurol India ; 72(2): 285-291, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38691471

ABSTRACT

BACKGROUND: Microparticles (MPs) have been implicated in thrombosis and endothelial dysfunction. Their involvement in early coagulopathy and in worsening of outcomes in isolated severe traumatic brain injury (sTBI) patients remains ill defined. OBJECTIVE: We sought to quantify the circulatory MP subtypes derived from platelets (PMPs; CD42), endothelial cells (EMPs; CD62E), and those bearing tissue factor (TFMP; CD142) and analyze their correlation with early coagulopathy, thrombin generation, and in-hospital mortality. MATERIALS AND METHODS: Prospective screening of sTBI patients was done. Blood samples were collected before blood and fluid transfusion. MP enumeration and characterization were performed using flow cytometry, and thrombin-antithrombin complex (TAT) levels were determined using enzyme-linked immunosorbent assay (ELISA). Circulating levels of procoagulant MPs were compared between isolated sTBI patients and age- and gender-matched healthy controls (HC). Patients were stratified according to their PMP, EMP, and TFMP levels, respectively (high ≥HC median and low < HC median). RESULTS: Isolated sTBI resulted in an increased generation of PMPs (456.6 [228-919] vs. 249.1 [198.9-404.5]; P = 0.01) and EMPs (301.5 [118.8-586.7] vs. 140.9 [124.9-286]; P = 0.09) compared to HCs. Also, 5.3% of MPs expressed TF (380 [301-710]) in HCs, compared to 6.6% MPs (484 [159-484]; P = 0.87) in isolated sTBI patients. Early TBI-associated coagulopathy (TBI-AC) was seen in 50 (41.6%) patients. PMP (380 [139-779] vs. 523.9 [334-927]; P = 0.19) and EMP (242 [86-483] vs. 344 [168-605]; P = 0.81) counts were low in patients with TBI-AC, compared to patients without TBI-AC. CONCLUSION: Our results suggest that enhanced cellular activation and procoagulant MP generation are predominant after isolated sTBI. TBI-AC was associated with low plasma PMPs count compared to the count in patients without TBI-AC. Low PMPs may be involved with the development of TBI-AC.


Subject(s)
Blood Coagulation Disorders , Brain Injuries, Traumatic , Cell-Derived Microparticles , Humans , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/mortality , Cell-Derived Microparticles/metabolism , Female , Male , Adult , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/blood , Middle Aged , Prospective Studies , Thromboplastin/metabolism , Blood Platelets/metabolism , Hospital Mortality , Endothelial Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...