1
|
Zhou ZP, Zhong L, Liu Y, Yang ZJ, Huang JJ, Li DZ, Chen YH, Luan YY, Yao YM, Wu M. Impact of early heparin therapy on mortality in critically ill patients with sepsis associated acute kidney injury: a retrospective study from the MIMIC-IV database. Front Pharmacol 2024; 14:1261305. [PMID: 38273840 PMCID: PMC10808568 DOI: 10.3389/fphar.2023.1261305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Background: Inflammatory-coagulation dysfunction plays an increasingly important role in sepsis associated acute kidney injury (SAKI). This study aimed to investigate whether early heparin therapy improves survival in patients with SAKI. Methods: Patients with SAKI were identified from the Medical Information Mart for Intensive Care-IV database. The patients were divided into two groups: those who received heparin subcutaneously within 48 h after intensive care unit (ICU) admission and the control group, who received no heparin. The primary endpoint was ICU mortality, the secondary outcomes were 7-day, 14-day, 28-day, and hospital mortality. Propensity score matching (PSM), marginal structural Cox model (MSCM), and E-value analyses were performed. Results: The study included 5623 individuals with SAKI, 2410 of whom received heparin and 3213 of whom did not. There were significant effects on ICU and 28-day mortality in the overall population with PSM. MSCM further reinforces the efficacy of heparin administration reduces ICU mortality in the general population. Stratification analysis with MSCM showed that heparin administration was associated with decreased ICU mortality at various AKI stages. Heparin use was also associated with reduced 28-day mortality in patients with only female, age >60 years, and AKI stage 3, with HRs of 0.79, 0.77, and 0.60, respectively (p < 0.05). E-value analysis suggests robustness to unmeasured confounding. Conclusion: Early heparin therapy for patients with SAKI decreased ICU mortality. Further analysis demonstrated that heparin therapy was associated with reduced 28-day mortality rate in patients only among female, age > 60 years and AKI stage 3.
Collapse
Affiliation(s)
- Zhi-Peng Zhou
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Li Zhong
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Guizhou University of Chinese Medicine, Guiyang, China
| | - Yan Liu
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Nosocomial Infection Prevention and Control, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Zhen-Jia Yang
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Postgraduate Education, Shantou University Medical College, Shantou, China
| | - Jia-Jia Huang
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Da-Zheng Li
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yu-Hua Chen
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Emergency Medicine, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Ying-Yi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yong-Ming Yao
- Trauma Research Center, Medical Innovation Research Department and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Ming Wu
- Department of Infection and Critical Care Medicine, Health Science Center, Shenzhen Second People’s Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Nosocomial Infection Prevention and Control, Shenzhen Second People’s Hospital, Shenzhen, China
- Department of Emergency Medicine, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
2
|
Liu J, Xie Y, Shu X, Chen Y, Sun Y, Zhong K, Liang H, Li Y, Yang C, Han Y, Zou Y, Zhuyi Z, Huang J, Li J, Hu X, Yi B. Value function assessment to different RL algorithms for heparin treatment policy of patients with sepsis in ICU. Artif Intell Med 2024; 147:102726. [PMID: 38184357 DOI: 10.1016/j.artmed.2023.102726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 01/08/2024]
Abstract
Heparin is a critical aspect of managing sepsis after abdominal surgery, which can improve microcirculation, protect organ function, and reduce mortality. However, there is no clinical evidence to support decision-making for heparin dosage. This paper proposes a model called SOFA-MDP, which utilizes SOFA scores as states of MDP, to investigate clinic policies. Different algorithms provide different value functions, making it challenging to determine which value function is more reliable. Due to ethical restrictions, we cannot test all policies on patients. To address this issue, we proposed two value function assessment methods: action similarity rate and relative gain. We experimented with heparin treatment policies for sepsis patients after abdominal surgery using MIMIC-IV. In the experiments, TD(0) shows the most reliable performance. Using the action similarity rate and relative gain to assess AI policy from TD(0), the agreement rates between AI policy and "good" physician's actual treatment are 64.6% and 73.2%, while the agreement rates between AI policy and "bad" physician's actual treatment are 44.1% and 35.8%, the gaps are 20.5% and 37.4%, respectively. External validation using action similarity rate and relative gain based on eICU resulted in agreement rates of 61.5% and 69.1% with the "good" physician's treatment, and 45.2% and 38.3% with the "bad" physician's treatment, with gaps of 16.3% and 30.8%, respectively. In conclusion, the model provides instructive support for clinical decisions, and the evaluation methods accurately distinguish reliable and unreasonable outcomes.
Collapse
Affiliation(s)
- Jiang Liu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China.
| | - Yihao Xie
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Xin Shu
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yuwen Chen
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Yizhu Sun
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Kunhua Zhong
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Hao Liang
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yujie Li
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Chunyong Yang
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yan Han
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yuwei Zou
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Ziting Zhuyi
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jiahao Huang
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Junhong Li
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoyan Hu
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Bin Yi
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
3
|
Alhazmi AS. Anticoagulative activity of Commiphora gileadensis, aspirin, and heparin on blood coagulation profiles in naïve mice. Int J Health Sci (Qassim) 2024; 18:10-16. [PMID: 38188900 PMCID: PMC10768467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Objective Commiphora gileadensis is a small tree under the genus Commiphora. Previous studies showed medical applications, such as antibacterial and antihypertensive, for C. gileadensis. Methods Sixty naïve mice were classified into six groups: control, C. gileadensis sap-treated group, C. gileadensis methanol extract-treated group, C. gileadensis acetone extract-treated group, heparin-treated group, and aspirin-treated group. Blood samples from each mouse in the six groups were collected in EDTA, sodium citrate, and heparin tubes. The body weight of each mouse was measured at the beginning and end of the experiment. Furthermore, complete blood count, kidney and renal function tests, coagulation profiles, prothrombin time (PT), activated partial thromboplastin time (aPTT), international normalized ratio (INR), D-dimer, and fibrinogen concentrations were estimated for each mouse. Results The sodium, potassium, chloride, blood urea nitrogen, creatinine, alanine transaminase, and aspartate transaminase levels did not show statistical differences between all groups. Moreover, PT, aPTT, and INR were prolonged in the C. gileadensis sap, methanol, and acetone extracts-treated mice compared with those in the heparin and aspirin-treated groups (P < 0.01). D-dimer and fibrinogen concentrations did not show significant statistical differences between all groups. Conclusion The current study concludes that the C. gileadensis sap, methanol, and acetone extracts prolonged PT, aPTT, and bleeding time in naïve mice more than heparin and aspirin. This means that the C. gileadensis extracts may have antithrombotic activity and may be used in the future to resolve intravascular thrombosis in patients having prosthetic valves.
Collapse
Affiliation(s)
- Ayman Saeed Alhazmi
- Department of Clinical Biochemistry, Applied Medical Sciences, Taif University, Saudi Arabia
| |
Collapse
|
4
|
van Solinge TS, Friis KP, O'Brien K, Verschoor RL, van Aarle J, Koekman A, Breakefield XO, Vader P, Schiffelers R, Broekman M. Heparin interferes with the uptake of liposomes in glioma. Int J Pharm X 2023; 6:100191. [PMID: 37408568 PMCID: PMC10319201 DOI: 10.1016/j.ijpx.2023.100191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
In glioblastoma, a malignant primary brain tumor, liposomes have shown promise in pre-clinical and early phase clinical trials as delivery vehicles for therapeutics. However, external factors influencing cellular uptake of liposomes in glioma cells are poorly understood. Heparin and heparin analogues are commonly used in glioma patients to decrease the risk of thrombo-embolic events. Our results show that heparin inhibits pegylated liposome uptake by U87 glioma and GL261 cells in a dose dependent manner in vitro, and that heparin-mediated inhibition of uptake required presence of fetal bovine serum in the media. In a subcutaneous model of glioma, Cy5.5 labeled liposomes could be detected with in vivo imaging after direct intra-tumoral injection. Ex-vivo analysis with flow cytometry showed a decreased uptake of liposomes into tumor cells in mice treated systemically with heparin compared to those treated with vehicle only.
Collapse
Affiliation(s)
- Thomas S. van Solinge
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
- Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Kristina Pagh Friis
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Killian O'Brien
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Romy L. Verschoor
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Jeroen van Aarle
- Department of Neurosurgery, University Medical Center, Utrecht, the Netherlands
| | - Arnold Koekman
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Xandra O. Breakefield
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Raymond Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marike Broekman
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
- Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands
- Department of Neurosurgery, Haaglanden Medical Center, The Hague, the Netherlands
| |
Collapse
|
5
|
Medeiros SK, Sharma N, Dwivedi D, Cani E, Zhou J, Dwivedi N, Sohrabipour S, Liaw PC. THE EFFECTS OF DNASE I AND LOW-MOLECULAR-WEIGHT HEPARIN IN A MURINE MODEL OF POLYMICROBIAL ABDOMINAL SEPSIS. Shock 2023; 59:666-672. [PMID: 36852972 DOI: 10.1097/shk.0000000000002095] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
ABSTRACT Introduction: Cell-free DNA (CFDNA) has emerged as a prognostic biomarker in patients with sepsis. Circulating CFDNA is hypothesized to be associated with histones in the form of nucleosomes. In vitro, DNA activates coagulation and inhibits fibrinolysis, whereas histones activate platelets and are cytotoxic to endothelial cells. Previous studies have targeted CFDNA or histones in animal models of sepsis using DNase I or heparins, respectively, which has reduced inflammatory and thrombosis markers, thereby improving survival. In this study, we explored the possibility that the combination of DNase I and a low-molecular weight heparin (LMWH) may be a better therapeutic approach than monotherapy in a murine model of abdominal sepsis. Methods: C57Bl/6 mice (8-12 weeks old, both sexes) were subjected to either cecal ligation and puncture or sham surgery. Mice were given antibiotics, fluids, and either saline, DNase I (intraperitoneally, 20 mg/kg/8 h), LMWH (dalteparin, subcutaneously 500 IU/kg/12 h), or a combination of both (n = 12-31). Mice were monitored over 72 h for survival. Organs and blood were harvested for analysis. Levels of LMWH, CFDNA, IL-6, citrullinated histone-H3, thrombin-antithrombin complexes, and protein C were measured in plasma. Results: Administration of either DNase I (81.8%) or LMWH (83.3%, prophylactic range of 0.12 ± 0.07 IU/mL achieved) improved the survival of septic mice compared with saline- (38.7%) and combination-treated mice (48.8%, P < 0.05). Combination-treated mice also showed a small but insignificant improvement in survival compared with saline-treated cecal ligation and puncture mice. Monotherapies may be improving survival by reducing blood bacterial loads, citrullinated histone-H3, and thrombin-antithrombin complexes, and improving protein C levels. Conclusions: Compared with saline- and combination-treated mice, administration of monotherapies to septic mice improved survival. These findings suggest that there may be a negative drug-drug interaction between DNase I and LMWH when DNase I is administered intraperitoneally in a murine model of polymicrobial abdominal sepsis.
Collapse
Affiliation(s)
| | | | | | | | | | - Naviya Dwivedi
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Canada
| | | | | |
Collapse
|
6
|
Zou ZY, Huang JJ, Luan YY, Yang ZJ, Zhou ZP, Zhang JJ, Yao YM, Wu M. Early prophylactic anticoagulation with heparin alleviates mortality in critically ill patients with sepsis: a retrospective analysis from the MIMIC-IV database. BURNS & TRAUMA 2022; 10:tkac029. [PMID: 36168402 PMCID: PMC9501718 DOI: 10.1093/burnst/tkac029] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/14/2022] [Indexed: 02/05/2023]
Abstract
Background Minimal data exist on anticoagulation use and timing and the dose of heparin in patients with sepsis, and whether heparin use improves sepsis survival remains largely unclear. This study was performed to assess whether heparin administration would provide a survival advantage in critically ill patients with sepsis. Methods A retrospective cohort study of patients with sepsis in the Medical Information Mart for Intensive Care (MIMIC)-IV database was conducted. Cox proportional hazards model and propensity score matching (PSM) were used to evaluate the outcomes of prophylactic anticoagulation with heparin administered by subcutaneous injection within 48 h of intensive care unit (ICU) admission. The primary outcome was in-hospital mortality. Secondary outcomes included 60-day mortality, length of ICU stay, length of hospital stay and incidence of acute kidney injury (AKI) on day 7. E-Value analysis were used for unmeasured confounding. Results A total of 6646 adult septic patients were included and divided into an early prophylactic heparin group (n = 3211) and a nonheparin group (n = 3435). In-hospital mortality in the heparin therapy group was significantly lower than that in the nonheparin group (prematched 14.7 vs 20.0%, hazard ratio (HR) 0.77, 95% confidence interval (CI) [0.68-0.87], p < 0.001, and postmatched 14.9 vs 18.3%, HR 0.78, 95% CI [0.68-0.89], p < 0.001). Secondary endpoints, including 60-day mortality and length of ICU stay, differed between the heparin and nonheparin groups (p < 0.01). Early prophylactic heparin administration was associated with in-hospital mortality among septic patients in different adjusted covariates (HR 0.71-0.78, p < 0.001), and only administration of five doses of heparin was associated with decreased in-hospital mortality after PSM (HR 0.70, 95% CI 0.56-0.87, p < 0.001). Subgroup analysis showed that heparin use was significantly associated with reduced in-hospital mortality in patients with sepsis-induced coagulopathy, septic shock, sequential organ failure assessment score ≥ 10, AKI, mechanical ventilation, gram-positive bacterial infection and gram-negative bacterial infection, with HRs of 0.74, 0.70, 0.58, 0.74, 0.73, 0.64 and 0.72, respectively (p <0.001). E-Value analysis suggested robustness to unmeasured confounding. Conclusions This study found an association between early administration prophylactic heparin provided to patients with sepsis and reduced risk-adjusted mortality. A prospective randomized-controlled study should be designed to further assess the relevant findings.
Collapse
Affiliation(s)
- Zhi-ye Zou
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Jia-jia Huang
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
- Postgraduate Education, Shantou University Medical College, Shantou 515041, China
| | - Ying-yi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Zhen-jia Yang
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
- Postgraduate Education, Shantou University Medical College, Shantou 515041, China
| | - Zhi-peng Zhou
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Jing-jing Zhang
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
- Postgraduate Education, Shantou University Medical College, Shantou 515041, China
| | - Yong-ming Yao
- Trauma Research Center, Medical Innovation Research Department and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, China
| | - Ming Wu
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
- Postgraduate Education, Shantou University Medical College, Shantou 515041, China
- Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
7
|
Yang R, Zhang X. A potential new pathway for heparin treatment of sepsis-induced lung injury: inhibition of pulmonary endothelial cell pyroptosis by blocking hMGB1-LPS-induced caspase-11 activation. Front Cell Infect Microbiol 2022; 12:984835. [PMID: 36189354 PMCID: PMC9519888 DOI: 10.3389/fcimb.2022.984835] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
Sepsis is a significant cause of mortality in critically ill patients. Acute lung injury (ALI) is a leading cause of death in these patients. Endothelial cells exposed to the bacterial endotoxin lipopolysaccharide (LPS) can progress into pyroptosis, a programmed lysis of cell death triggered by inflammatory caspases. It is characterized by lytic cell death induced by the binding of intracellular LPS to caspases 4/5 in human cells and caspase-11 in mouse cells. In mice,caspase-11-dependent pyroptosis plays an important role in endotoxemia. HMGB1 released into the plasma binds to LPS and is internalized into lysosomes in endothelial cells via the advanced glycation end product receptor. In the acidic lysosomal environment, HMGB1 permeates the phospholipid bilayer, which is followed by the leakage of LPS into the cytoplasm and the activation of caspase-11. Heparin is an anticoagulant widely applied in the treatment of thrombotic disease. Previous studies have found that heparin could block caspase-11-dependent inflammatory reactions, decrease sepsis-related mortality, and reduce ALI, independent of its anticoagulant activity. Heparin or modified heparin with no anticoagulant property could inhibit the alarmin HMGB1-LPS interactions, minimize LPS entry into the cytoplasm, and thus blocking caspase-11 activation. Heparin has been studied in septic ALI, but the regulatory mechanism of pulmonary endothelial cell pyroptosis is still unclear. In this paper, we discuss the potential novel role of heparin in the treatment of septic ALI from the unique mechanism of pulmonary endothelial cell pyroptosis.
Collapse
|
8
|
Nording H, Baron L, Haberthür D, Emschermann F, Mezger M, Sauter M, Sauter R, Patzelt J, Knoepp K, Nording A, Meusel M, Meyer-Saraei R, Hlushchuk R, Sedding D, Borst O, Eitel I, Karsten CM, Feil R, Pichler B, Erdmann J, Verschoor A, Chavakis E, Chavakis T, von Hundelshausen P, Köhl J, Gawaz M, Langer HF. The C5a/C5a receptor 1 axis controls tissue neovascularization through CXCL4 release from platelets. Nat Commun 2021; 12:3352. [PMID: 34099640 PMCID: PMC8185003 DOI: 10.1038/s41467-021-23499-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 03/28/2021] [Indexed: 02/05/2023] Open
Abstract
Platelets contribute to the regulation of tissue neovascularization, although the specific factors underlying this function are unknown. Here, we identified the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) on platelets as a negative regulatory mechanism of vessel formation. We showed that platelets expressing C5aR1 exert an inhibitory effect on endothelial cell functions such as migration and 2D and 3D tube formation. Growth factor- and hypoxia-driven vascularization was markedly increased in C5ar1-/- mice. Platelet-specific deletion of C5aR1 resulted in a proangiogenic phenotype with increased collateralization, capillarization and improved pericyte coverage. Mechanistically, we found that C5a induced preferential release of CXC chemokine ligand 4 (CXCL4, PF4) from platelets as an important antiangiogenic paracrine effector molecule. Interfering with the C5aR1-CXCL4 axis reversed the antiangiogenic effect of platelets both in vitro and in vivo.In conclusion, we identified a mechanism for the control of tissue neovascularization through C5a/C5aR1 axis activation in platelets and subsequent induction of the antiangiogenic factor CXCL4.
Collapse
Affiliation(s)
- Henry Nording
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany ,grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Lasse Baron
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - David Haberthür
- grid.5734.50000 0001 0726 5157Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Frederic Emschermann
- grid.10392.390000 0001 2190 1447University Hospital, Department of Cardiovascular Medicine, Eberhard Karls University, Tübingen, Germany
| | - Matthias Mezger
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Manuela Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Reinhard Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Johannes Patzelt
- grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Kai Knoepp
- grid.9018.00000 0001 0679 2801Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Halle (Saale), Germany
| | - Anne Nording
- grid.10392.390000 0001 2190 1447Institute of Medical Genetics and Applied Genomics, Eberhard Karls University, Tübingen, Germany
| | - Moritz Meusel
- grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Roza Meyer-Saraei
- grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Ruslan Hlushchuk
- grid.5734.50000 0001 0726 5157Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Daniel Sedding
- grid.9018.00000 0001 0679 2801Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Halle (Saale), Germany
| | - Oliver Borst
- grid.10392.390000 0001 2190 1447University Hospital, Department of Cardiovascular Medicine, Eberhard Karls University, Tübingen, Germany
| | - Ingo Eitel
- grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Christian M. Karsten
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Robert Feil
- grid.10392.390000 0001 2190 1447Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Bernd Pichler
- grid.10392.390000 0001 2190 1447Institute for Preclinical Imaging, Eberhard Karls University, Tübingen, Germany
| | - Jeanette Erdmann
- grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.4562.50000 0001 0057 2672Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Admar Verschoor
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Emmanouil Chavakis
- grid.411088.40000 0004 0578 8220Department for Internal Medicine III/Cardiology, University Hospital of the Johann-Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Triantafyllos Chavakis
- grid.4488.00000 0001 2111 7257Department of Clinical Pathobiochemistry, Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Philipp von Hundelshausen
- grid.5252.00000 0004 1936 973XInstitute for Cardiovascular Prevention, Ludwig Maximilians University Munich, Munich, Germany
| | - Jörg Köhl
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany ,grid.239573.90000 0000 9025 8099Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Meinrad Gawaz
- grid.10392.390000 0001 2190 1447University Hospital, Department of Cardiovascular Medicine, Eberhard Karls University, Tübingen, Germany
| | - Harald F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany ,grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| |
Collapse
|
9
|
Tang Y, Wang X, Li Z, He Z, Yang X, Cheng X, Peng Y, Xue Q, Bai Y, Zhang R, Zhao K, Liang F, Xiao X, Andersson U, Wang H, Billiar TR, Lu B. Heparin prevents caspase-11-dependent septic lethality independent of anticoagulant properties. Immunity 2021; 54:454-467.e6. [PMID: 33561388 DOI: 10.1016/j.immuni.2021.01.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 11/09/2020] [Accepted: 01/13/2021] [Indexed: 01/01/2023]
Abstract
Heparin, a mammalian polysaccharide, is a widely used anticoagulant medicine to treat thrombotic disorders. It is also known to improve outcomes in sepsis, a leading cause of mortality resulted from infection-induced immune dysfunction. Whereas it is relatively clear how heparin exerts its anticoagulant effect, the immunomodulatory mechanisms enabled by heparin remain enigmatic. Here, we show that heparin prevented caspase-11-dependent immune responses and lethality in sepsis independent of its anticoagulant properties. Heparin or a chemically modified form of heparin without anticoagulant function inhibited the alarmin HMGB1-lipopolysaccharide (LPS) interaction and prevented the macrophage glycocalyx degradation by heparanase. These events blocked the cytosolic delivery of LPS in macrophages and the activation of caspase-11, a cytosolic LPS receptor that mediates lethality in sepsis. Survival was higher in septic patients treated with heparin than those without heparin treatment. The identification of this previously unrecognized heparin function establishes a link between innate immune responses and coagulation.
Collapse
Affiliation(s)
- Yiting Tang
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, 410000 P.R. China
| | - Xiangyu Wang
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Zhaozheng Li
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Zhihui He
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Xinyu Yang
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Xiaoye Cheng
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Yue Peng
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Qianqian Xue
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Yang Bai
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Rui Zhang
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Kai Zhao
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Fang Liang
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China
| | - Xianzhong Xiao
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, 410000 P.R. China; Key Laboratory of sepsis translational medicine of Hunan, Central South University, Changsha, Hunan Province, 410000 P.R. China
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, 171 76, Stockholm, Sweden
| | - Haichao Wang
- The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213 USA
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, 410000 P.R. China; Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, 410000 P.R. China; Key Laboratory of sepsis translational medicine of Hunan, Central South University, Changsha, Hunan Province, 410000 P.R. China.
| |
Collapse
|
10
|
Pearce L, Davidson SM, Yellon DM. The cytokine storm of COVID-19: a spotlight on prevention and protection. Expert Opin Ther Targets 2020; 24:723-730. [PMID: 32594778 DOI: 10.1080/14728222.2020.1783243] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The cytokine release syndrome (CRS) of COVID-19 is associated with the development of critical illness requiring multi-organ support. Further research is required to halt progression of multi-organ injury induced by hyper-inflammation. AREAS COVERED PubMed/MEDLINETM databases were accessed between May 9th-June 9th, 2020, to review the latest perspectives on the treatment and pathogenesis of CRS. EXPERT OPINION Over-activity of chemotaxis triggers a macrophage activation syndrome (MAS) resulting in the release of pro-inflammatory cytokines. IL-6 and TNF- α are at the forefront of hyper-inflammation. The inflammatory cascade induces endothelial activation and capillary leak, leading to circulatory collapse and shock. As endothelial dysfunction persists, there is activation of the clotting cascade and microvascular obstruction. Continued endothelial activation results in multi-organ failure, regardless of pulmonary tissue damage. We propose that targeting the endothelium may interrupt this cycle. Immuno-modulating therapies have been suggested, however, further data is necessary to confirm that they do not jeopardize adaptive immunity. Inhibition of IL-6 and the Janus Kinase, signal transducer and activator of transcription proteins pathway (JAK/STAT), are favorable targets. Remote ischemic conditioning (RIC) reduces the inflammation of sepsis in animal models and should be considered as a low risk intervention, in combination with cardiovascular protection.
Collapse
Affiliation(s)
- Lucie Pearce
- The Hatter Cardiovascular Institute, University College London , London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London , London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London , London, UK
| |
Collapse
|
11
|
Camprubí-Rimblas M, Tantinyà N, Guillamat-Prats R, Bringué J, Puig F, Gómez MN, Blanch L, Artigas A. Effects of nebulized antithrombin and heparin on inflammatory and coagulation alterations in an acute lung injury model in rats. J Thromb Haemost 2020; 18:571-583. [PMID: 31755229 PMCID: PMC9906372 DOI: 10.1111/jth.14685] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/29/2019] [Accepted: 11/18/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND During acute respiratory distress syndrome, proinflammatory mediators inhibit natural anticoagulant factors, which alter the normal balance between coagulation and fibrinolysis leading to a procoagulant state. We hypothesize that pulmonary administration of anticoagulants might be beneficial to treat acute respiratory distress syndrome for their anticoagulant and antiinflammatory effects and reduce the risk of systemic bleeding. OBJECTIVES Our aim is to study the effects of nebulized antithrombin (AT) and combined AT and heparin in an animal model of acute lung injury. METHODS Acute lung injury was induced in rats by the intratracheal administration of hydrochloric acid and lipopolysaccharide. AT alone (500 IU/kg body weight) or combined with heparin (1000 IU/kg body weight) were nebulized after the injury. Control groups received saline instead. Blood, lung tissue, bronchoalveolar lavage, and alveolar macrophages (AM) isolated from bronchoalveolar lavage were collected after 48 hours and analyzed. RESULTS Nebulized anticoagulant treatments reduced protein concentration in the lungs and decreased injury-mediated coagulation factors (tissue factor, plasminogen activator inhibitor-1, plasminogen, and fibrinogen degradation product) and inflammation (tumor necrosis factor α and interleukin 1β) in the alveolar space without affecting systemic coagulation and no bleeding. AT alone reduced fibrin deposition and edema in the lungs. Heparin did not potentiate AT coagulant effect but promoted the reduction of macrophages infiltration into the alveolar compartment. Anticoagulants reduced nuclear factor-kB downstream effectors in AM. CONCLUSIONS Nebulized AT and heparin attenuate lung injury through decreasing coagulation and inflammation without altering systemic coagulation and no bleeding. However, combined AT and heparin did not produce a synergistic effect.
Collapse
Affiliation(s)
- Marta Camprubí-Rimblas
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Neus Tantinyà
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Raquel Guillamat-Prats
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Josep Bringué
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ferranda Puig
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | | | - Lluís Blanch
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Critical Care Center, Corporació Sanitària i Universitària Parc Taulí, Sabadell, Spain
| | - Antonio Artigas
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Critical Care Center, Corporació Sanitària i Universitària Parc Taulí, Sabadell, Spain
| |
Collapse
|
12
|
Lê BV, Jandrot-Perrus M, Couture C, Checkmahomed L, Venable MC, Hamelin MÈ, Boivin G. Evaluation of anticoagulant agents for the treatment of human metapneumovirus infection in mice. J Gen Virol 2018; 99:1367-1380. [PMID: 30102144 DOI: 10.1099/jgv.0.001135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Thrombin has been demonstrated to be involved in several viral diseases including human metapneumovirus (hMPV) infections. We previously showed that immediate administration of thrombin inhibitor argatroban post-infection protected mice against hMPV disease. This current work aims at determining whether warfarin and heparin, two other anticoagulants inhibiting thrombin formation and activities, may also be used for treatment against hMPV in vivo. We found that immediate injections of argatroban, warfarin or heparin after virus challenge protected mice against hMPV infection, as evidenced by decreased or no mortality, less weight loss, reduced viral load and attenuated inflammation. However, delayed treatments starting 1 day post-infection with argatroban or warfarin almost did not impact the survival whereas delayed treatment with heparin induced an increased mortality during infection. Moreover, these treatments also did not reduce weight loss, viral replication and inflammation. In agreement with these results, thrombin generation was decreased upon immediate anticoagulant treatments but was unaltered upon delayed treatments. Thus, thrombin generation occurs at the onset of hMPV infection and thrombin inhibition may be only useful for the treatment of this disease when initiated in the early stage. In this case, heparin is not recommended because of its reduced efficacy on mortality in infected mice whereas argatroban and warfarin appear as safe and effective drugs for the treatment of hMPV disease. The antiviral and anti-inflammatory effects of argatroban occur via thrombin-dependent pathways whereas the mechanisms by which warfarin exerts its beneficial effects against hMPV infection were not elucidated and need to be further studied.
Collapse
Affiliation(s)
- Ba Vuong Lê
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Christian Couture
- 3Quebec Heart and Lung Institute, Laval University, Quebec City, Quebec, Canada
| | - Liva Checkmahomed
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Marie-Ève Hamelin
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Guy Boivin
- 1Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
13
|
Dao DT, Anez-Bustillos L, Ourieff J, Pan A, Mitchell PD, Kishikawa H, Fell GL, Baker MA, Watnick RS, Chen H, Hamilton TE, Rogers MS, Bielenberg DR, Puder M. Heparin impairs angiogenic signaling and compensatory lung growth after left pneumonectomy. Angiogenesis 2018; 21:837-848. [PMID: 29956017 DOI: 10.1007/s10456-018-9628-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/26/2018] [Indexed: 12/24/2022]
Abstract
Children with hypoplastic lung diseases, such as congenital diaphragmatic hernia, can require life support via extracorporeal membrane oxygenation and systemic anticoagulation, usually in the form of heparin. The role of heparin in angiogenesis and organ growth is inconclusive, with conflicting data reported in the literature. This study aimed to investigate the effects of heparin on lung growth in a model of compensatory lung growth (CLG). Compared to the absence of heparin, treatment with heparin decreased the vascular endothelial growth factor (VEGF)-mediated activation of VEGFR2 and mitogenic effect on human lung microvascular endothelial cells in vitro. Compared to non-heparinized controls, heparinized mice demonstrated impaired pulmonary mechanics, decreased respiratory volumes and flows, and reduced activity levels after left pneumonectomy. They also had lower lung volume, pulmonary septal surface area and alveolar density on morphometric analyses. Lungs of heparinized mice displayed decreased phosphorylation of VEGFR2 compared to the control group, with consequential downstream reduction in markers of cellular proliferation and survival. The use of bivalirudin, an alternative anticoagulant that does not interact with VEGF, preserved lung growth and pulmonary mechanics. These results demonstrated that heparin impairs CLG by reducing VEGFR2 activation. These findings raise concern for the clinical use of heparin in the setting of organ growth or regeneration.
Collapse
Affiliation(s)
- Duy T Dao
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Lorenzo Anez-Bustillos
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Jared Ourieff
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Amy Pan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Paul D Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Hiroko Kishikawa
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Gillian L Fell
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Meredith A Baker
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Randolph S Watnick
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Thomas E Hamilton
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Michael S Rogers
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Diane R Bielenberg
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - Mark Puder
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA. .,Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Ahmed A, Dolasia K, Mukhopadhyay S. Mycobacterium tuberculosisPPE18 Protein Reduces Inflammation and Increases Survival in Animal Model of Sepsis. THE JOURNAL OF IMMUNOLOGY 2018; 200:3587-3598. [DOI: 10.4049/jimmunol.1602065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/14/2018] [Indexed: 12/16/2022]
|
15
|
Chimenti L, Camprubí-Rimblas M, Guillamat-Prats R, Gomez MN, Tijero J, Blanch L, Artigas A. Nebulized Heparin Attenuates Pulmonary Coagulopathy and Inflammation through Alveolar Macrophages in a Rat Model of Acute Lung Injury. Thromb Haemost 2017; 117:2125-2134. [PMID: 29202212 PMCID: PMC6328369 DOI: 10.1160/th17-05-0347] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective
Alveolar macrophages play a key role in the development and resolution of acute respiratory distress syndrome (ARDS), modulating the inflammatory response and the coagulation cascade in lungs. Anti-coagulants may be helpful in the treatment of ARDS. This study investigated the effects of nebulized heparin on the role of alveolar macrophages in limiting lung coagulation and inflammatory response in an animal model of acute lung injury (ALI).
Methods
Rats were randomized to four experimental groups. In three groups, ALI was induced by intratracheal instillation of lipopolysaccharide (LPS) and heparin was nebulized at constant oxygen flow: the LPS/Hep group received nebulized heparin 4 and 8 hours after injury; the Hep/LPS/Hep group received nebulized heparin 30 minutes before and 4 and 8 hours after LPS-induced injury; the LPS/Sal group received nebulized saline 4 and 8 hours after injury. The control group received only saline. Animals were exsanguinated 24 hours after LPS instillation. Lung tissue, bronchoalveolar lavage fluid (BALF) and alveolar macrophages isolated from BALF were analysed.
Results
LPS increased protein concentration, oedema and neutrophils in BALF as well as procoagulant and proinflammatory mediators in lung tissue and alveolar macrophages. In lung tissue, nebulized heparin attenuated ALI through decreasing procoagulant (tissue factor, thrombin–anti-thrombin complexes, fibrin degradation products) and proinflammatory (interleukin 6, tumour necrosis factor alpha) pathways. In alveolar macrophages, nebulized heparin reduced expression of procoagulant genes and the effectors of transforming growth factor beta (Smad 2, Smad 3) and nuclear factor kappa B (p-selectin, CCL-2). Pre-treatment resulted in more pronounced attenuation.
Conclusion
Nebulized heparin reduced pulmonary coagulopathy and inflammation without producing systemic bleeding, partly by modulating alveolar macrophages.
Collapse
Affiliation(s)
- Laura Chimenti
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain
| | - Marta Camprubí-Rimblas
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Raquel Guillamat-Prats
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Maria Nieves Gomez
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain
| | - Jessica Tijero
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain
| | - Lluis Blanch
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain.,Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Critical Care Center, Corporació Sanitària i Universitària Parc Taulí-UAB, Sabadell, Catalonia, Spain
| | - Antonio Artigas
- Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain.,Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Critical Care Center, Corporació Sanitària i Universitària Parc Taulí-UAB, Sabadell, Catalonia, Spain
| |
Collapse
|
16
|
Shashar M, Belghasem ME, Matsuura S, Walker J, Richards S, Alousi F, Rijal K, Kolachalama VB, Balcells M, Odagi M, Nagasawa K, Henderson JM, Gautam A, Rushmore R, Francis J, Kirchhofer D, Kolandaivelu K, Sherr DH, Edelman ER, Ravid K, Chitalia VC. Targeting STUB1-tissue factor axis normalizes hyperthrombotic uremic phenotype without increasing bleeding risk. Sci Transl Med 2017; 9:eaam8475. [PMID: 29167396 PMCID: PMC5854487 DOI: 10.1126/scitranslmed.aam8475] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 07/31/2017] [Accepted: 10/04/2017] [Indexed: 12/30/2022]
Abstract
Chronic kidney disease (CKD/uremia) remains vexing because it increases the risk of atherothrombosis and is also associated with bleeding complications on standard antithrombotic/antiplatelet therapies. Although the associations of indolic uremic solutes and vascular wall proteins [such as tissue factor (TF) and aryl hydrocarbon receptor (AHR)] are being defined, the specific mechanisms that drive the thrombotic and bleeding risks are not fully understood. We now present an indolic solute-specific animal model, which focuses on solute-protein interactions and shows that indolic solutes mediate the hyperthrombotic phenotype across all CKD stages in an AHR- and TF-dependent manner. We further demonstrate that AHR regulates TF through STIP1 homology and U-box-containing protein 1 (STUB1). As a ubiquitin ligase, STUB1 dynamically interacts with and degrades TF through ubiquitination in the uremic milieu. TF regulation by STUB1 is supported in humans by an inverse relationship of STUB1 and TF expression and reduced STUB1-TF interaction in uremic vessels. Genetic or pharmacological manipulation of STUB1 in vascular smooth muscle cells inhibited thrombosis in flow loops. STUB1 perturbations reverted the uremic hyperthrombotic phenotype without prolonging the bleeding time, in contrast to heparin, the standard-of-care antithrombotic in CKD patients. Our work refines the thrombosis axis (STUB1 is a mediator of indolic solute-AHR-TF axis) and expands the understanding of the interconnected relationships driving the fragile thrombotic state in CKD. It also establishes a means of minimizing the uremic hyperthrombotic phenotype without altering the hemostatic balance, a long-sought-after combination in CKD patients.
Collapse
Affiliation(s)
- Moshe Shashar
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mostafa E Belghasem
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Shinobu Matsuura
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Joshua Walker
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Sean Richards
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Faisal Alousi
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Keshab Rijal
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Vijaya B Kolachalama
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mercedes Balcells
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Biological Engineering Department, Institut Químic de Sarrià, Universitat Ramon Llull, Barcelona 08017, Spain
| | - Minami Odagi
- Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kazuo Nagasawa
- Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Amitabh Gautam
- Department of Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Richard Rushmore
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jean Francis
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Daniel Kirchhofer
- Department of Early Discovery and Biochemistry, Genentech Inc., South San Francisco, CA 94080, USA
| | - Kumaran Kolandaivelu
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David H Sherr
- Department of Environmental Health, School of Public Health, Boston University School of Medicine, Boston, MA 02118, USA
| | - Elazer R Edelman
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Vipul C Chitalia
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
17
|
Zhao D, Ding R, Liu Y, Yin X, Zhang Z, Ma X. Unfractionated heparin protects the protein C system against lipopolysaccharide-induced damage in vivo and in vitro. Exp Ther Med 2017; 14:5515-5522. [PMID: 29285085 DOI: 10.3892/etm.2017.5236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 07/14/2017] [Indexed: 01/27/2023] Open
Abstract
Activation of protein C is greatly enhanced by the presence of thrombomodulin (TM) and endothelial protein C receptor (EPCR) on the endothelial surface. Impairment of the anticoagulant protein C system occurs during endotoxemia and contributes to sepsis-associated hypercoagulability. Previous studies have demonstrated that unfractionated heparin (UFH) can attenuate coagulation in endotoxemic mice. However, whether UFH has an effect on the protein C system remains to be elucidated. The current study evaluated the therapeutic effect of UFH on the protein C system in a mouse model of lipopolysaccharide (LPS)-induced sepsis, and further investigated the effect of UFH on the expression of TM and EPCR in vitro using human umbilical vein endothelial cells (HUVECs). The in vivo data indicated that UFH preconditioning attenuated the decline in circulating activated protein C following LPS administration, and also reduced LPS-induced shedding of TM and EPCR. In HUVECs, LPS stimulation led to the downregulation of TM and EPCR expression, and UFH dose-dependently restored the mRNA and protein levels of TM and EPCR. In addition, UFH inhibited the LPS-induced activation of mitogen-activated protein kinase 14, proto-oncogene tyrosine-protein kinase Src and nuclear factor κB signaling in HUVECs. In summary, these results suggest that UFH has a protective effect on the protein C system during sepsis. Thus, UFH may be a candidate therapeutic agent for the treatment of patients with sepsis.
Collapse
Affiliation(s)
- Dongmei Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Renyu Ding
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yina Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaohan Yin
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhidan Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaochun Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
18
|
Artigas A, Camprubí-Rimblas M, Tantinyà N, Bringué J, Guillamat-Prats R, Matthay MA. Inhalation therapies in acute respiratory distress syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:293. [PMID: 28828368 DOI: 10.21037/atm.2017.07.21] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The defining features of acute respiratory distress syndrome (ARDS) are an excessive inflammatory respiratory response associated with high morbidity and mortality. Treatment consists mainly of measures to avoid worsening lung injury and cannot reverse the underlying pathophysiological process. New pharmacological agents have shown promising results in preclinical studies; however, they have not been successfully translated to patients with ARDS. The lack of effective therapeutic interventions has resulted in a recent interest in strategies to prevent ARDS with treatments delivering medications directly to the lungs by inhalation and nebulization, hopefully minimizing systemic adverse events. We analyzed the effect of different aerosolized drugs such as bronchodilators, corticosteroids, pulmonary vasodilators, anticoagulants, mucolytics and surfactant. New therapeutic strategies and ongoing trials using carbon monoxide (CO) and AP301 peptide are also briefly reviewed.
Collapse
Affiliation(s)
- Antonio Artigas
- Institut d'Investigació i Innovació Parc Tauli (I3PT), Sabadell, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Critical Care Center, Corporació Sanitària Universitaria Parc Taulí, Sabadell, Spain
| | - Marta Camprubí-Rimblas
- Institut d'Investigació i Innovació Parc Tauli (I3PT), Sabadell, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Neus Tantinyà
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Josep Bringué
- Institut d'Investigació i Innovació Parc Tauli (I3PT), Sabadell, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Raquel Guillamat-Prats
- Institut d'Investigació i Innovació Parc Tauli (I3PT), Sabadell, Spain.,Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Michael A Matthay
- Departments of Medicine and Anesthesia and Cardiovascular Research Institute, University of California, San Francisco, USA
| |
Collapse
|
19
|
Evans CE, Zhao YY. Impact of thrombosis on pulmonary endothelial injury and repair following sepsis. Am J Physiol Lung Cell Mol Physiol 2017; 312:L441-L451. [PMID: 28130261 PMCID: PMC5407094 DOI: 10.1152/ajplung.00441.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/20/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
The prevailing morbidity and mortality in sepsis are largely due to multiple organ dysfunction (MOD), most commonly lung injury, as well as renal and cardiac dysfunction. Despite recent advances in defining many aspects of the pathogenesis of sepsis-related MOD, including acute respiratory distress syndrome (ARDS), there are currently no effective pharmacological or cell-based treatments for the disease. Human and animal studies have shown that pulmonary thrombosis is common in sepsis-induced ARDS, and preclinical studies have shown that anticoagulation may improve outcome following sepsis challenge. The potential beneficial effect of anticoagulation on outcome is unconvincing in clinical studies, however, and these discrepancies may arise from the multiple and sometimes opposing actions of thrombosis on the pulmonary endothelium following sepsis. It has been suggested, for example, that mild pulmonary thrombosis prevents escape of bacterial infection into the circulation, while severe thrombosis causes hypoxia and results in pulmonary endothelial damage. Evidence from both human and animal studies has demonstrated the key role of microvascular leakage in determining the outcome of sepsis. In this review, we describe thrombosis-dependent mechanisms that regulate pulmonary endothelial injury and repair following sepsis, including activation of the coagulation cascade by tissue factor and stimulation of vascular repair by hypoxia-inducible factors. Targeting such mechanisms through anticoagulant, anti-inflammatory, and reparative methods may represent a novel approach for the treatment of septic patients.
Collapse
Affiliation(s)
- Colin E Evans
- Department of Pharmacology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois; and
- Center for Lung and Vascular Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois
| | - You-Yang Zhao
- Department of Pharmacology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois; and
- Center for Lung and Vascular Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois
| |
Collapse
|
20
|
Camprubí-Rimblas M, Guillamat-Prats R, Lebouvier T, Chimenti L, Iglesias M, Obiols C, Tijero J, Gómez MN, de Haro C, Blanch L, Artigas A. Heparin effect in alveolar cells and macrophages in an acute lung injury model. Intensive Care Med Exp 2015. [PMCID: PMC4796197 DOI: 10.1186/2197-425x-3-s1-a570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
21
|
Kuznetsova TA, Besednova NN, Somova LM, Plekhova NG. Fucoidan extracted from Fucus evanescens prevents endotoxin-induced damage in a mouse model of endotoxemia. Mar Drugs 2014; 12:886-98. [PMID: 24492521 PMCID: PMC3944521 DOI: 10.3390/md12020886] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/24/2014] [Accepted: 01/26/2014] [Indexed: 12/27/2022] Open
Abstract
An important problem of treating patients with endotoxemia is to find drugs to reduce the negative effects of endotoxin on the organism. We tested fucoidan (sulfated polysaccharide) from the brown alga Fucus evanescens as a potential drug in a mouse model of endotoxemia inducted by lipopolysaccharide (LPS). The survival time of mice injected with LPS increased under fucoidan treatment compared with the group of mice injected with LPS only. The preventive administration of fucoidan to mice with endotoxemia resulted in inhibition of increased levels of proinflammatory cytokines (TNFα and IL-6), as well as decreasing of the processes of hypercoagulability. The parenteral or per os administration of fucoidan resulted in decreasing the degree of microcirculatory disorders and secondary dystrophic-destructive changes in parenchymal organs of mice with endotoxemia. Taken together, these results demonstrate that fucoidan prevents endotoxin-induced damage in a mouse model of endotoxemia and increases the mice's resistance to LPS.
Collapse
Affiliation(s)
- Tatyana A Kuznetsova
- Laboratory of immunology, G.P. Somov Research Institute of Epidemiology and Microbiology, Siberian Branch of Russian Academy of Medical Sciences, 1 Selskya St., Vladivostok 690087, Russia.
| | - Natalya N Besednova
- Laboratory of immunology, G.P. Somov Research Institute of Epidemiology and Microbiology, Siberian Branch of Russian Academy of Medical Sciences, 1 Selskya St., Vladivostok 690087, Russia.
| | - Larisa M Somova
- Laboratory of pathomorphology, G.P. Somov Research Institute of Epidemiology and Microbiology, Siberian Branch of Russian Academy of Medical Sciences, 1 Selskya St., Vladivostok 690087, Russia.
| | - Natalya G Plekhova
- Laboratory of pathomorphology, G.P. Somov Research Institute of Epidemiology and Microbiology, Siberian Branch of Russian Academy of Medical Sciences, 1 Selskya St., Vladivostok 690087, Russia.
| |
Collapse
|
22
|
Qiu P, Li Y, Shiloach J, Cui X, Sun J, Trinh L, Kubler-Kielb J, Vinogradov E, Mani H, Al-Hamad M, Fitz Y, Eichacker PQ. Bacillus anthracis cell wall peptidoglycan but not lethal or edema toxins produces changes consistent with disseminated intravascular coagulation in a rat model. J Infect Dis 2013; 208:978-89. [PMID: 23737601 DOI: 10.1093/infdis/jit247] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Disseminated intravascular coagulation (DIC) appears to be important in the pathogenesis of Bacillus anthracis infection, but its causes are unclear. Although lethal toxin (LT) and edema toxin (ET) could contribute, B. anthracis cell wall peptidoglycan (PGN), not the toxins, stimulates inflammatory responses associated with DIC. METHODS AND RESULTS To better understand the pathogenesis of DIC during anthrax, we compared the effects of 24-hour infusions of PGN, LT, ET, or diluent (control) on coagulation measures 6, 24, or 48 hours after infusion initiation in 135 rats. No control recipient died. Lethality rates (approximately 30%) did not differ among PGN, LT, and ET recipients (P = .78). Thirty-three of 35 deaths (94%) occurred between 6 and 24 hours after the start of challenge. Among challenge components, PGN most consistently altered coagulation measures. Compared with control at 6 hours, PGN decreased platelet and fibrinogen levels and increased prothrombin and activated partial thromboplastin times and tissue factor, tissue factor pathway inhibitor, protein C, plasminogen activator inhibitor (PAI), and thrombin-antithrombin complex levels, whereas LT and ET only decreased the fibrinogen level or increased the PAI level (P ≤ .05). Nearly all effects associated with PGN infusion significantly differed from changes associated with toxin infusion (P ≤ .05 for all comparisons except for PAI level). CONCLUSION DIC during B. anthracis infection may be related more to components such as PGN than to LT or ET.
Collapse
Affiliation(s)
- Ping Qiu
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Borgel D, Lerolle N. Quel avenir pour les médicaments de l’hémostase dans le traitement du sepsis sévère après le Xigris® ? MEDECINE INTENSIVE REANIMATION 2013. [DOI: 10.1007/s13546-013-0665-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
24
|
Oudemans-van Straaten HM, Ostermann M. Bench-to-bedside review: Citrate for continuous renal replacement therapy, from science to practice. Crit Care 2012; 16:249. [PMID: 23216871 PMCID: PMC3672558 DOI: 10.1186/cc11645] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
To prevent clotting in the extracorporeal circuit during continuous renal replacement therapy (CRRT) anticoagulation is required. Heparin is still the most commonly used anticoagulant. However, heparins increase the risk of bleeding, especially in critically ill patients. Evidence has accumulated that regional anticoagulation of the CRRT circuit with citrate is feasible and safe. Compared to heparin, citrate anticoagulation reduces the risk of bleeding and requirement for blood products, not only in patients with coagulopathy, but also in those without. Metabolic complications are largely prevented by the use of a strict protocol, comprehensive training and integrated citrate software. Recent studies indicate that citrate can even be used in patients with significant liver disease provided that monitoring is intensified and the dose is carefully adjusted. Since the citric acid cycle is oxygen dependent, patients at greatest risk of accumulation seem to be those with persistent lactic acidosis due to poor tissue perfusion. The use of citrate may also be associated with less inflammation due to hypocalcemia-induced suppression of intracellular signaling at the membrane and avoidance of heparin, which may have proinflammatory properties. Whether these beneficial effects increase patient survival needs to be confirmed. However, other benefits are the reason that citrate should become the first choice anticoagulant for CRRT provided that its safe use can be guaranteed.
Collapse
Affiliation(s)
| | - Marlies Ostermann
- King's College London, Guy's and St Thomas' Hospital, Department of Critical Care and Nephrology, London SE1 7EH, UK
| |
Collapse
|
25
|
A comparison of the effect of convection against diffusion in hemodynamics and cytokines clearance in an experimental model of septic shock. J Trauma Acute Care Surg 2012; 73:855-60. [DOI: 10.1097/ta.0b013e31825eea8a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
26
|
|
27
|
|