1
|
Xoay TD, Tuan TA, Ha NT, Quan TQ, Duyen NT, My TTK. Antithrombin and Activated Protein C in Pediatric Sepsis: Prospective Observational Study of Outcome. Pediatr Crit Care Med 2025; 26:e197-e205. [PMID: 39718419 DOI: 10.1097/pcc.0000000000003677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
OBJECTIVES To assess antithrombin and activated protein C (aPC) levels in relation to disseminated intravascular coagulation (DIC) and severe outcomes in pediatric sepsis. DESIGN Prospective, observational study conducted between April 2023 and October 2024. Coagulation profiles including conventional coagulation, antithrombin activity, and aPC were obtained at PICU admission. SETTING PICU in the Vietnam National Children's Hospital, Hanoi, Vietnam. SUBJECTS PICU admissions, 1 month to 18 years old, with sepsis. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS One hundred thirty children (78 males; median age 7.5 mo) with mortality 23/130 (17.7%). The prevalence of overt DIC was 37 of 130 (28.5%). Nonsurvival at 28 days, compared with survival, was associated with hemorrhage and/or thrombosis at presentation, and higher number of dysfunctional organs, and overt DIC. Those with overt DIC, compared with not, had longer activated partial thromboplastin time, higher international normalized ratio and d -dimer, and lower antithrombin, and aPC. Activity of antithrombin and aPC correlated inversely with the Vasoactive-Inotropic Score in survivors ( p = 0.002 and 0.009, respectively). Patients with a cutoff value for antithrombin less than 63.5% had a mortality risk with area under the receiver operating characteristic (AUROC) curve 0.64, with sensitivity 0.51 and specificity 0.74, and positive predictive value 0.30. Regarding overt DIC, a cutoff value for antithrombin less than 55.5% had an AUROC 0.78, sensitivity 0.72 and specificity of 0.73, and positive predictive value 0.52. CONCLUSIONS In this observational study of pediatric sepsis patients, first 24-hour coagulation data in those who did not-survive to 28 days, vs. survivors showed an associated prior lower level of antithrombin in nonsurvivors. Furthermore, using the outcome of overt DIC and nonovert DIC in the first 72 hours, we found that lower levels of antithrombin or aPC are each associated with overt DIC and nonovert DIC in pediatric sepsis. Further validation work is needed in larger case series of pediatric sepsis.
Collapse
Affiliation(s)
- Tran Dang Xoay
- Hanoi Medical University, Hanoi, Vietnam
- Pediatric Intensive Care Unit, Vietnam National Children's Hospital, Hanoi, Vietnam
| | - Ta Anh Tuan
- Hanoi Medical University, Hanoi, Vietnam
- Pediatric Intensive Care Unit, Vietnam National Children's Hospital, Hanoi, Vietnam
| | | | - Thieu Quang Quan
- Pediatric Intensive Care Unit, Vietnam National Children's Hospital, Hanoi, Vietnam
| | - Nguyen Thi Duyen
- Department of Hematology, Vietnam National Children's Hospital, Hanoi, Vietnam
| | | |
Collapse
|
2
|
Man C, An Y, Wang GX, Mao EQ, Ma L. Recent Advances in Pathogenesis and Anticoagulation Treatment of Sepsis-Induced Coagulopathy. J Inflamm Res 2025; 18:737-750. [PMID: 39845020 PMCID: PMC11752821 DOI: 10.2147/jir.s495223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025] Open
Abstract
Coagulopathy in sepsis is common and is associated with high mortality. Although immunothrombosis is necessary for infection control, excessive thrombus formation can trigger a systemic thrombo-inflammatory response. Immunothrombosis plays a core role in sepsis-induced coagulopathy, and research has revealed a complex interplay between inflammation and coagulation. Different mechanisms underlying sepsis-related coagulopathy are discussed, including factors contributing to the imbalance of pro- and anticoagulation relevant to endothelial cells. The potential therapeutic implications of anticoagulants on these mechanisms are discussed. This review contributes to our understanding of the pathogenesis of coagulopathy in patients with sepsis. Recent studies suggest that endothelial cells play an important role in immunoregulation and hemostasis. Meanwhile, the non-anticoagulation effects of anticoagulants, especially heparin, which act in the pathogenesis of coagulopathy in septic patients, have been partially revealed. We believe that further insights into the pathogenesis of sepsis-induced coagulopathy will help physicians evaluate patient conditions effectively, leading to advanced early recognition and better decision-making in the treatment of sepsis.
Collapse
Affiliation(s)
- Chit Man
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Yuan An
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Guo-Xin Wang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - En-Qiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Li Ma
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
3
|
Sharew SG, Weldehanna DG, Gebreyes DS, Abebe TA, Shibabaw A. Extended spectrum betalactamase and Carbapenemase producing gram negative bacteria from healthcare workers gowns at Debre Berhan Comprehensive Specialized Hospital, Ethiopia. Sci Rep 2025; 15:1784. [PMID: 39805904 PMCID: PMC11729860 DOI: 10.1038/s41598-024-84563-1] [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: 08/11/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
Extended-spectrum beta-lactamase (ESBL) and carbapenemase-producing (CP) gram-negative bacteria are the major public health concerns. Gowns used by healthcare workers (HCWs) in daily practice are a source of hospital-acquired infections in hospital settings. The study aimed to determine the prevalence of extended-spectrum beta-lactamase and carbapenemase-producing gram-negative bacteria from gowns of healthcare workers at Debre Berhan Comprehensive Specialized Hospital, Amhara Regional State, Ethiopia. A hospital-based cross-sectional study was conducted from August to October 2022. A total of 226 swab samples were collected from gowns of HCWs and cultured on MacConkey agar. Bacterial identifications were done by standard biochemical tests. Screening for ESBL and carbapenemase production was done using CHROME agar. Confirmation of ESBL and carbapenemase production was done by the combination disk method and modified carbapenem inactivation method, respectively. Data were analyzed by using SPSS version 25. The overall contamination rate of gowns was 46.9% (106/226). Among 226 swab samples, a total of 117 (51.7%) gram-negative bacteria were isolated. Among these, the most frequent isolates were E. coli, accounted 36 (30.8%) followed by K. pneumoniae, 26(22.2%). The overall multi-drug resistance (MDR) rate was 65 (55.6%). Of the 117 isolates, 17(14.5%) and 12(10.3%) were ESBL and carbapenemase producers respectively. Gown type (p = 0.041), laundering practice (p = 0.045), number of gowns (p = 0.002), and gown washing frequency per week (p = 0.017).were significantly associated with bacterial contamination of gowns. In this study, the prevalence of ESBL and CP Gram-negative bacteria from gowns of healthcare workers was found to be alarming. Therefore, strict infection prevention and control practices, as well as good hygienic practices, should be implemented to reduce and prevent cross-contamination and the spread of antimicrobial-resistant strains in hospital settings.
Collapse
Affiliation(s)
- Sisay Gebrish Sharew
- Department of Medical Laboratory Science, Debere Birhan Health Science College. Debre, Berhan, Ethiopia
| | - Daniel Gebretsadik Weldehanna
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Demissew Shenkute Gebreyes
- Department of Medical Laboratory Science, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia.
| | - Tsegahun Asfaw Abebe
- Department of Medical Laboratory Science, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia
| | - Agumas Shibabaw
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| |
Collapse
|
4
|
Mukhtar O, Lal A, Jentzer J, Kashani K. Validation of SCAI Shock Staging in Critically Ill Medical Intensive Care Unit Patients With Sepsis and Septic Shock. J Community Hosp Intern Med Perspect 2025; 15:13-21. [PMID: 39867142 PMCID: PMC11759074 DOI: 10.55729/2000-9666.1436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 01/28/2025] Open
Abstract
Purpose This study evaluated the predictive value of SCAI shock staging for mortality in patients with sepsis and septic shock admitted to the medical ICU. Materials and methods This is a single-center historical cohort study. We analyzed data for adults (≥18-year-old) admitted to the medical ICU at Mayo Clinic St. Mary's campus with sepsis between June 1, 2018, and December 31, 2021. Sepsis was identified using the Sepsis-III criteria. Patients were stratified based on SCAI shock staging. Our primary outcome was all-cause 30-day mortality. Results We identified 3079 eligible adult patients with sepsis or septic shock. The distribution of SCAI shock stages A through E was 9%, 12%, 25%, 49%, and 5%, respectively. The overall 30-day mortality was 24%. There was progression in all outcomes including ICU, hospital and 30-day mortality across SCAI shock stages. However, only SCAI shock stages D and E, had statistically significant adjusted HRs of 1.6 and 3, respectively. When compared to SOFA score, SCAI shock staging performed similarly in predicting ICU mortality with no statistically significant difference in AUCs, p-value of 0.07. Conclusions Our results support the use of SCAI shock staging in critically ill medical patients with sepsis and septic shock for risk stratification. We propose that the SCAI shock staging may be used as a universal system for grading the severity of shock in critically ill patients regardless of etiology.
Collapse
Affiliation(s)
- Osama Mukhtar
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN,
USA
| | - Amos Lal
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN,
USA
| | - Jacob Jentzer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN,
USA
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN,
USA
| |
Collapse
|
5
|
Ninivaggi M, Sokolova L, Donkervoort D, de Laat B, de Laat-Kremers R. Thrombomodulin is a stronger indicator of combined oral contraceptives-induced activated protein C pathway resistance in the thrombin generation test than activated protein C. Front Cardiovasc Med 2024; 11:1490601. [PMID: 39677039 PMCID: PMC11638229 DOI: 10.3389/fcvm.2024.1490601] [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: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Background The mechanism by which combined oral contraceptives (COCs) lead to hypercoagulation is not fully understood, although activated protein C (APC) pathway resistance has been implicated. APC and thrombomodulin (TM) tend to be considered as interchangeable reagents, even though their biological action in coagulation is different. However, it remains unclear which reagent is better suited for the detection of APC pathway resistance. We compared the effectiveness of TM and APC in TG to detect COC-induced APC pathway resistance using thrombin generation (TG). Methods TG was measured on ST Genesia in 48 healthy women, of whom 24 used COCs. TG was triggered with STG-ThromboScreen (with and without TM), spiked with a low and high concentration of TM or APC (2 or 15 nM TM, or 1.5 or 5.5 nM APC), aimed to achieve 50% and 90% ETP inhibition, respectively. Results TG was higher in women using COCs. TM and APC inhibit TG in all women, although their inhibitory effect is more pronounced in women without COC compared to women with COC. The addition of 2 nM TM causes an ETP reduction of 40% (1,289 vs. 768 nM•min) in women without COC and an ETP reduction of 24% (1,704 vs. 1,287 nM•min) in women with COC. The addition of 1.5 nM APC causes an ETP reduction of 41% (1,289 vs. 759 nM•min) in women without COC and an ETP reduction of 23% (1,704 vs. 1,316 nM•min) in women with COC. The difference in effect between women with and without COC is largest when 15 nM TM, aimed at 90% ETP inhibition, is used. 15 nM TM leads to the smallest overlap in ETP inhibition between women with and without COC (27% overlap), compared to 2 nM TM (41% overlap), and 1.5 nM APC (38% overlap) and 5.5 nM APC (41% overlap). Conclusion Although TM and APC are often used interchangeably to assess the sensitivity of the APC system in TG, our findings suggest that TM is a better discriminator to detect COC-use induced APC pathway resistance. In addition, we found that the ETP is a better TG test readout for APC pathway resistance testing than the peak height.
Collapse
Affiliation(s)
- Marisa Ninivaggi
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, Netherlands
| | - Lily Sokolova
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, Netherlands
- Department of Data Analysis and Artificial Intelligence, Synapse Research Institute, Maastricht, Netherlands
| | - Demy Donkervoort
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, Netherlands
| | - Bas de Laat
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, Netherlands
- Department of Data Analysis and Artificial Intelligence, Synapse Research Institute, Maastricht, Netherlands
| | - Romy de Laat-Kremers
- Department of Data Analysis and Artificial Intelligence, Synapse Research Institute, Maastricht, Netherlands
| |
Collapse
|
6
|
Yong J, Toh CH. The convergent model of coagulation. J Thromb Haemost 2024; 22:2140-2146. [PMID: 38815754 DOI: 10.1016/j.jtha.2024.05.014] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
It is increasingly apparent that the pathologic interplay between coagulation and innate immunity, ie, immunothrombosis, forms the common basis of many challenges across the boundaries of specialized medicine and cannot be fully explained by the conventional concepts of cascade and cell-based coagulation. To improve our understanding of coagulation, we propose a model of coagulation that converges with inflammation and innate immune activation as a unified response toward vascular injury. Evolutionarily integral to the convergent response are damage-associated molecular patterns, which are released as a consequence of injury. Damage-associated molecular patterns facilitate diverse interactions within and between systems, not only to complement and reinforce cell-based clot formation but also to steer the response toward clot resolution and wound healing. By extending coagulation beyond its current boundaries, the convergent model aims to deliver novel diagnostics and therapeutics for contemporary and unexpected challenges across medicine, as exposed by COVID-19 and vaccine-induced immune thrombotic thrombocytopenia.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
7
|
Eichinger M, Rief M, Eichlseder M, Pichler A, Zoidl P, Hallmann B, Zajic P. Hyperkalaemia in bleeding trauma patients: A potential marker of disease severity - A retrospective cohort study. Heliyon 2024; 10:e30037. [PMID: 38765110 PMCID: PMC11101751 DOI: 10.1016/j.heliyon.2024.e30037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 05/21/2024] Open
Abstract
Background Hyperkalaemia is a common electrolyte abnormality seen in critically ill patients. In haemorrhagic shock, it may contribute to cardiac arrest and has been identified as a potential marker for tissue hypoxia. However, the significance of its role in haemorrhagic shock and its contribution to mortality remains unclear. This study aimed to examine the potential underlying pathophysiology and evaluate the incidence and characteristics of patients with hyperkalaemia on hospital arrival in bleeding trauma patients before transfusions and its mortality. Methods A retrospective cohort study was conducted on adult patients with traumatic bleeding admitted to a European Major Trauma Centre between January 2016 and December 2021. Patients were classified according to their serum potassium levels on arrival, and relevant clinical parameters between non-hyperkalaemic and hyperkalaemic patients were compared. Results Among the 83 patients in this study, 8 (9.6 %) presented with hyperkalaemia on arrival. The median shock index showed a higher tendency in the hyperkalaemic group. Hyperkalaemia was found to be more common among younger patients who sustained penetrating trauma. Mortality rates were higher in the hyperkalaemic group, but the difference was not statistically significant. Conclusion Our results suggest that hyperkalaemia occurs frequently in bleeding trauma patients on hospital arrival pre-transfusions, indicating a more severe illness. Our findings provide insights into the pathophysiology and characteristics of hyperkalaemia in bleeding trauma patients. Further studies are required to investigate the mechanisms by which hyperkalaemia contributes to mortality in haemorrhagic shock patients.
Collapse
Affiliation(s)
- Michael Eichinger
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| | - Martin Rief
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| | - Michael Eichlseder
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| | - Alexander Pichler
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| | - Philipp Zoidl
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| | - Barbara Hallmann
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| | - Paul Zajic
- Division of Anaesthesiology and Intensive Care Medicine 1, Medical University of Graz, Graz, Austria
| |
Collapse
|
8
|
Williams B, Zou L, Pittet JF, Chao W. Sepsis-Induced Coagulopathy: A Comprehensive Narrative Review of Pathophysiology, Clinical Presentation, Diagnosis, and Management Strategies. Anesth Analg 2024; 138:696-711. [PMID: 38324297 PMCID: PMC10916756 DOI: 10.1213/ane.0000000000006888] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Physiological hemostasis is a balance between pro- and anticoagulant pathways, and in sepsis, this equilibrium is disturbed, resulting in systemic thrombin generation, impaired anticoagulant activity, and suppression of fibrinolysis, a condition termed sepsis-induced coagulopathy (SIC). SIC is a common complication, being present in 24% of patients with sepsis and 66% of patients with septic shock, and is often associated with poor clinical outcomes and high mortality. 1 , 2 Recent preclinical and clinical studies have generated new insights into the molecular pathogenesis of SIC. In this article, we analyze the complex pathophysiology of SIC with a focus on the role of procoagulant innate immune signaling in hemostatic activation--tissue factor production, thrombin generation, endotheliopathy, and impaired antithrombotic functions. We also review clinical presentations of SIC, the diagnostic scoring system and laboratory tests, the current standard of care, and clinical trials evaluating the efficacies of anticoagulant therapies.
Collapse
Affiliation(s)
- Brittney Williams
- From the Division of Cardiothoracic Anesthesia, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Jean-Francois Pittet
- Division of Critical Care, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Johri N, Matreja PS, Agarwal S, Nagar P, Kumar D, Maurya A. Unraveling the Molecular Mechanisms of Activated Protein C (APC) in Mitigating Reperfusion Injury and Cardiac Ischemia: a Promising Avenue for Novel Therapeutic Interventions. J Cardiovasc Transl Res 2024; 17:345-355. [PMID: 37851312 DOI: 10.1007/s12265-023-10445-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Ischemic heart disease, which results from plaque formation in the coronary arteries, hinders the flow of oxygenated blood to the heart, leading to ischemia. Reperfusion injury remains a significant challenge for researchers, and the mechanisms underlying myocardial ischemia-reperfusion injury (MIRI) are not entirely understood. The review directs future research into potential targets in clinical treatment based on our present understanding of the pathophysiological mechanisms of MIRI. The study provides insights into the mechanisms underlying MIRI and offers direction for future research in this area. The use of targeted therapies may hold promise in improving cardiac function in the elderly and minimizing the adverse effects of revascularization therapies. The purpose of this review is to analyze the role of activated protein C (APC) in the pathogenesis of ischemic heart disease, heart failure, and myocardial ischemia-reperfusion injury, and discuss the potential of APC-based therapeutics.
Collapse
Affiliation(s)
- Nishant Johri
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India.
- School of Health & Psychological Sciences, City, University of London, London, United Kingdom.
| | - Prithpal S Matreja
- Department of Pharmacology, Teerthanker Mahaveer Medical College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Shalabh Agarwal
- Department of Cardiology, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Priya Nagar
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Deepanshu Kumar
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Aditya Maurya
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| |
Collapse
|
10
|
Borsellino B, Bravo-Perez C, Visconte V, Guarnera L. Thrombosis in Myeloid Malignancies: From CHIP to AML. Cardiovasc Hematol Disord Drug Targets 2024; 24:2-12. [PMID: 38879768 DOI: 10.2174/011871529x307253240530060107] [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: 01/29/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 09/04/2024]
Abstract
The development of myeloid malignancies is a multi-step process starting from pre-malignant stages. Large-scale studies on clonal hematopoiesis of indeterminate potential (CHIP) identified this condition as a risk factor for developing hematologic malignancies, in particular myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). In parallel, CHIP was found to confer an enhanced thrombotic risk, in particular for cardiovascular diseases. In a similar fashion, in recent years, alongside their life-threatening features, increasing attention has been drawn toward thrombotic complications in myeloid malignancies. Thus, the purpose of this review is to gather a growing body of evidence on incidence, pathogenesis and clinical impact of thrombosis in myeloid malignancies at every step of malignant progression, from CHIP to AML.
Collapse
Affiliation(s)
- Beatrice Borsellino
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Carlos Bravo-Perez
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH44195, USA
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER-Instituto de Salud Carlos III, 30005, Murcia, Spain
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH44195, USA
| | - Luca Guarnera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH44195, USA
| |
Collapse
|
11
|
Palomo M, Moreno-Castaño AB, Salas MQ, Escribano-Serrat S, Rovira M, Guillen-Olmos E, Fernandez S, Ventosa-Capell H, Youssef L, Crispi F, Nomdedeu M, Martinez-Sanchez J, De Moner B, Diaz-Ricart M. Endothelial activation and damage as a common pathological substrate in different pathologies and cell therapy complications. Front Med (Lausanne) 2023; 10:1285898. [PMID: 38034541 PMCID: PMC10682735 DOI: 10.3389/fmed.2023.1285898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
The endothelium is a biologically active interface with multiple functions, some of them common throughout the vascular tree, and others that depend on its anatomical location. Endothelial cells are continually exposed to cellular and humoral factors, and to all those elements (biological, chemical, or hemodynamic) that circulate in blood at a certain time. It can adapt to different stimuli but this capability may be lost if the stimuli are strong enough and/or persistent in time. If the endothelium loses its adaptability it may become dysfunctional, becoming a potential real danger to the host. Endothelial dysfunction is present in multiple clinical conditions, such as chronic kidney disease, obesity, major depression, pregnancy-related complications, septic syndromes, COVID-19, and thrombotic microangiopathies, among other pathologies, but also in association with cell therapies, such as hematopoietic stem cell transplantation and treatment with chimeric antigen receptor T cells. In these diverse conditions, evidence suggests that the presence and severity of endothelial dysfunction correlate with the severity of the associated disease. More importantly, endothelial dysfunction has a strong diagnostic and prognostic value for the development of critical complications that, although may differ according to the underlying disease, have a vascular background in common. Our multidisciplinary team of women has devoted many years to exploring the role of the endothelium in association with the mentioned diseases and conditions. Our research group has characterized some of the mechanisms and also proposed biomarkers of endothelial damage. A better knowledge would provide therapeutic strategies either to prevent or to treat endothelial dysfunction.
Collapse
Affiliation(s)
- Marta Palomo
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Hematology External Quality Assessment Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Ana Belén Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| | - María Queralt Salas
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, Barcelona, Spain
| | - Silvia Escribano-Serrat
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, Barcelona, Spain
| | - Elena Guillen-Olmos
- Department of Nephrology and Kidney Transplantation, Hospital Clínic de Barcelona, Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud (CSUR), University of Barcelona, Barcelona, Spain
| | - Sara Fernandez
- Medical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Lina Youssef
- BCNatal – Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic de Barcelona and Hospital Sant Joan de Déu, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Fatima Crispi
- BCNatal – Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic de Barcelona and Hospital Sant Joan de Déu, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Meritxell Nomdedeu
- Hemostasis and Hemotherapy Department, Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Maribel Diaz-Ricart
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Yuan L, Han J, van der Velden AIM, Vink H, de Mutsert R, Rosendaal FR, van Hylckama Vlieg A, Li-Gao R, Rabelink TJ, van den Berg BM. Sex-specific association between microvascular health and coagulation parameters: the Netherlands Epidemiology of Obesity study. J Thromb Haemost 2023; 21:2585-2595. [PMID: 37301258 DOI: 10.1016/j.jtha.2023.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/16/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Microvascular dysfunction is a growing determinant of sex differences in coronary heart disease (CHD). Dysregulation of the coagulation system is involved in CHD pathogenesis and can be induced by endothelial glycocalyx (EG) perturbation. However, little is known about the link between EG function and coagulation parameters in population-based studies on sex specificity. OBJECTIVES We sought to examine the sex differences in the relationship between EG function and coagulation parameters in a middle-aged Dutch population. METHODS Using baseline measurements of 771 participants from the Netherlands Epidemiology of Obesity study (age, 56 years [IQR, 51-61 years]; 53% women; body mass index, 27.9 kg/m2 [IQR, 25.1-30.9 kg/m2]), associations between glycocalyx-related perfused boundary region (PBR) derived using sidestream dark-field imaging and coagulation parameters (factor [F]VIII/IX/XI; thrombin generation parameters; and fibrinogen) were investigated using linear regression analyses, adjusting for possible confounders (including C-reactive protein, leptin, and glycoprotein acetyls), followed by sex-stratified analyses. RESULTS There was a sex difference in the associations between PBR and coagulation parameters. Particularly in women, 1-SD PBR (both total and feed vessel, indicating poorer glycocalyx status) was associated with higher FIX activity ([1.8%; 95% CI, 0.3%-3.3%] and [2.0%; 95% CI, 0.5%-3.4%], respectively) and plasma fibrinogen levels ([5.1 mg/dL; 95% CI, 0.4-9.9 mg/dL] and [5.8 mg/dL; 95% CI, 1.1-10.6 mg/dL], respectively). Furthermore, 1-SD PBRcapillary was associated with higher FVIII activity (3.5%; 95% CI, 0.4%-6.5%) and plasma fibrinogen levels (5.3 mg/dL; 95% CI, 0.6-10.0 mg/dL). CONCLUSION We revealed a sex-specific association between microcirculatory health and procoagulant status, which suggests that microvascular health be considered during early development of CHD in women.
Collapse
Affiliation(s)
- Lushun Yuan
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jihee Han
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anouk I M van der Velden
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans Vink
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; MicroVascular Health Solutions LLC, Alpine, Utah, USA
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands; Metabolon Inc, Morrisville, North Carolina, USA
| | - Ton J Rabelink
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bernard M van den Berg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
13
|
Jang JH, Shin KH, Lee HR, Son E, Lee SE, Seol HY, Yoon SH, Kim T, Cho WH, Jeon D, Kim YS, Yeo HJ. Initial Tumor Necrosis Factor-Alpha and Endothelial Activation Are Associated with Hemorrhagic Complications during Extracorporeal Membrane Oxygenation. J Clin Med 2023; 12:4520. [PMID: 37445555 DOI: 10.3390/jcm12134520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Studies on inflammatory markers, endothelial activation, and bleeding during extracorporeal membrane oxygenation (ECMO) are lacking. Blood samples were prospectively collected after ECMO initiation from 150 adult patients who underwent ECMO for respiratory failure between 2018 and 2021. After excluding patients who died early (within 48 h), 132 patients were finally included. Their tumor necrosis factor-alpha (TNF-α), tissue factor (TF), soluble thrombomodulin (sTM), and E-selectin levels were measured. A Cox proportional hazards regression model was used to estimate the hazard ratio for hemorrhagic complications during ECMO. The 132 patients were divided into hemorrhagic (n = 23, H group) and non-complication (n = 109, N group) groups. The sequential organ failure assessment score, hemoglobin level, and ECMO type were included as covariates in all Cox models to exclude the effects of clinical factors. After adjusting for these factors, initial TNF-α, TF, sTM, E-selectin, and activated protein C levels were significantly associated with hemorrhagic complications (all p < 0.001). TNF-α, TF, and E-selectin better predicted hemorrhagic complications than the model that included only the aforementioned clinical factors (clinical factors only (area under the curve [AUC]: 0.804), reference; TNF-α (AUC: 0.914); TF (AUC: 0.915); E-selectin (AUC: 0.869)). Conclusions: TNF-α levels were significantly predictive of hemorrhagic complications during ECMO.
Collapse
Affiliation(s)
- Jin Ho Jang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Kyung-Hwa Shin
- Department of Laboratory Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Hye Rin Lee
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Eunjeong Son
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Seung Eun Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Hee Yun Seol
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Seong Hoon Yoon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Taehwa Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Woo Hyun Cho
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Doosoo Jeon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Yun Seong Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Hye Ju Yeo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Transplantation Research Center and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| |
Collapse
|
14
|
Hermsen J, Hambley B. The Coagulopathy of Acute Promyelocytic Leukemia: An Updated Review of Pathophysiology, Risk Stratification, and Clinical Management. Cancers (Basel) 2023; 15:3477. [PMID: 37444587 DOI: 10.3390/cancers15133477] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Acute promyelocytic leukemia (APL) has a well-established mechanism and a long-term prognosis that exceeds that of any other acute leukemia. These improving outcomes are due, in part, to all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), two targeted and highly active agents in this disease. However, there remains a considerable morbidity and mortality risk in APL secondary to clinically significant hemorrhagic and/or thrombotic events. Prevention and treatment of these coagulopathic complications remain significant impediments to further progress in optimizing outcomes for patients with APL. Moreover, the relative rarity of APL hinders adequately powered randomized controlled trials for evaluating APL coagulopathy management strategies. This review draws from peer-reviewed works falling between initial descriptions of APL in 1957 and work published prior to January 2023 and provides an updated overview of the pathophysiology of hemorrhagic and thrombotic complications in APL, outlines risk stratification parameters, and compiles current clinical best practices. An improved understanding of the pathophysiologic mechanisms driving hemorrhage and thrombosis along with the completion of well-designed trials of management strategies will assist clinicians in developing interventions that mitigate these devastating complications in an otherwise largely curable disease.
Collapse
Affiliation(s)
- Jack Hermsen
- University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Bryan Hambley
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, 3125 Eden Ave, Cincinnati, OH 45267, USA
| |
Collapse
|
15
|
Cánovas-Cervera I, Nacher-Sendra E, Osca-Verdegal R, Dolz-Andrés E, Beltrán-García J, Rodríguez-Gimillo M, Ferrando-Sánchez C, Carbonell N, García-Giménez JL. The Intricate Role of Non-Coding RNAs in Sepsis-Associated Disseminated Intravascular Coagulation. Int J Mol Sci 2023; 24:ijms24032582. [PMID: 36768905 PMCID: PMC9916911 DOI: 10.3390/ijms24032582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Disseminated Intravascular Coagulation (DIC) is a type of tissue and organ dysregulation in sepsis, due mainly to the effect of the inflammation on the coagulation system. Unfortunately, the underlying molecular mechanisms that lead to this disorder are not fully understood. Moreover, current biomarkers for DIC, including biological and clinical parameters, generally provide a poor diagnosis and prognosis. In recent years, non-coding RNAs have been studied as promising and robust biomarkers for a variety of diseases. Thus, their potential in the diagnosis and prognosis of DIC should be further studied. Specifically, the relationship between the coagulation cascade and non-coding RNAs should be established. In this review, microRNAs, long non-coding RNAs, and circular RNAs are studied in relation to DIC. Specifically, the axis between these non-coding RNAs and the corresponding affected pathway has been identified, including inflammation, alteration of the coagulation cascade, and endothelial damage. The main affected pathway identified is PI3K/AKT/mTOR axis, where several ncRNAs participate in its regulation, including miR-122-5p which is sponged by circ_0005963, ciRS-122, and circPTN, and miR-19a-3p which is modulated by circ_0000096 and circ_0063425. Additionally, both miR-223 and miR-24 were found to affect the PI3K/AKT pathway and were regulated by lncGAS5 and lncKCNQ1OT1, respectively. Thus, this work provides a useful pipeline of inter-connected ncRNAs that future research on their impact on DIC can further explore.
Collapse
Affiliation(s)
- Irene Cánovas-Cervera
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Health Research Institute INCLIVA, 46010 Valencia, Spain
| | - Elena Nacher-Sendra
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Health Research Institute INCLIVA, 46010 Valencia, Spain
| | - Rebeca Osca-Verdegal
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Research Network on Rare Diseases (CIBERER), Carlos III Health Institute, 46010 Valencia, Spain
| | - Enric Dolz-Andrés
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Jesús Beltrán-García
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Research Network on Rare Diseases (CIBERER), Carlos III Health Institute, 46010 Valencia, Spain
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, CA 92093, USA
| | - María Rodríguez-Gimillo
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - Carolina Ferrando-Sánchez
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - Nieves Carbonell
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - José Luis García-Giménez
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Research Network on Rare Diseases (CIBERER), Carlos III Health Institute, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-963-864-646
| |
Collapse
|
16
|
Lian H, Zhang H, Ding X, Wang X. The importance of a sepsis layered early warning system for critical patients. Am J Transl Res 2022; 14:5229-5242. [PMID: 36105025 PMCID: PMC9452367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/12/2022] [Indexed: 06/15/2023]
Abstract
Critical illness, particularly sepsis, is associated with high mortality, so prevention is more important than effective therapy. Advances in medical science have provided more opportunities for early warning and early intervention to avoid the development of critical illness. Existing early warning systems (EWS) have the advantages of high efficiency and convenience. However, with the development of medical technology, they do not completely meet clinical needs. EWS should contain elements that meet many dimensions of clinical requirements, including risk warning, response warning, injury warning, critical warning, and death warning. By summarizing previous studies, we outlined a layered EWS that follows RISK bundles. RISK represents different warning sign categories: R: host response, I: organ injury, S: changes in vital signs, and K: gradual appearance of "killed" organs. We plan to construct a complete layered EWS to guide clinical activities and subsequent clinical studies in the near future.
Collapse
Affiliation(s)
- Hui Lian
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, P. R. China
| | - Hongmin Zhang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, P. R. China
| | - Xin Ding
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, P. R. China
| | - Xiaoting Wang
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, P. R. China
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, P. R. China
| |
Collapse
|
17
|
Saadi S, Allem R, Sebaihia M, Merouane A, Bakkali M. Bacterial contamination of neglected hospital surfaces and equipment in an Algerian hospital: an important source of potential infection. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:1373-1381. [PMID: 33648396 DOI: 10.1080/09603123.2021.1885631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 01/30/2021] [Indexed: 06/12/2023]
Abstract
Hospital surfaces are heavily contaminated with bacteria, which are a potential source of nosocomial infections. This study was undertaken to identify bacterial communities isolated from neglected hospital surfaces after cleaning routine in a Algerian public hospital. Screening of bacterial contamination in patient bed (PB), reception land-line phones (TF), door handles (DH) and medical equipment (ME) during five months generated 108 inocula. Isolates obtained were identified based on biochemical characteristics and confirmed by analysis of 16S rRNA sequences. Statistical analysis was performed to reveal possible relationship between bacterial diversity and swabbed surfaces. Our findings showed a high prevalence of bacteria in various hospital surfaces, reaching (65.25%), where a highest contaminated surface was the PB (47.22%) and a lowest contaminated was TF (5.55%). Gram negative bacteria were the dominant group (62.03%) mainly represented by Entrobacteriaceae (42.59%), whereas Staphylococcus aureus belonging to Gram positive was the main expanded pathogen with (15.74%).
Collapse
Affiliation(s)
- Somia Saadi
- Laboratory of Molecular Biology, Genomics and Bioinformatics, Department of Biology, Faculty of Natural and Life Sciences, Hassiba Benbouali University, Chlef, Algeria
| | - Rachida Allem
- Laboratory of Natural Bioresources, Faculty of Nature and Life Sciences, Hassiba Benbouali University, Chlef, Algeria
| | - Mohammed Sebaihia
- Laboratory of Molecular Biology, Genomics and Bioinformatics, Department of Biology, Faculty of Natural and Life Sciences, Hassiba Benbouali University, Chlef, Algeria
| | - Abdelaziz Merouane
- Laboratory of Natural Bioresources, Faculty of Nature and Life Sciences, Hassiba Benbouali University, Chlef, Algeria
| | - Mohammed Bakkali
- Departamento de Genetica, Facultad de Ciencias, Universidad de Granada Fuentenueva S/N, 18071, Granada, Spain
| |
Collapse
|
18
|
Dufour-Gaume F, Javelle E, Sailliol A, Cap AP, Prat NJ. COVID-19 : viral infection, endotheliopathy and the immuno-inflammatory response… is it time to consider a standard (non-immunized) plasma therapy approach to maintain homeostasis? Transfus Clin Biol 2022; 29:191-194. [PMID: 35644840 PMCID: PMC9059338 DOI: 10.1016/j.tracli.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | - Emilie Javelle
- Hôpital d'Instruction des Armées Laveran, Marseille, France
| | - Anne Sailliol
- Institut de Recherche Biomédicale des Armées, France
| | - Andre P Cap
- U.S. Army Institute of Surgical Research, Houston, USA
| | | |
Collapse
|
19
|
Zhu L, Gao T, Huang Y, Jin J, Wang D, Zhang L, Jin Y, Li P, Hu Y, Wu Y, Liu H, Dong Q, Wang G, Zheng T, Song C, Bai Y, Zhang X, Liu Y, Yang W, Xu K, Zou G, Zhao L, Cao R, Zhong W, Xia X, Xiao G, Liu X, Cao C. Ebola virus VP35 hijacks the PKA-CREB1 pathway for replication and pathogenesis by AKIP1 association. Nat Commun 2022; 13:2256. [PMID: 35474062 PMCID: PMC9042921 DOI: 10.1038/s41467-022-29948-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/06/2022] [Indexed: 12/17/2022] Open
Abstract
Ebola virus (EBOV), one of the deadliest viruses, is the cause of fatal Ebola virus disease (EVD). The underlying mechanism of viral replication and EBOV-related hemorrhage is not fully understood. Here, we show that EBOV VP35, a cofactor of viral RNA-dependent RNA polymerase, binds human A kinase interacting protein (AKIP1), which consequently activates protein kinase A (PKA) and the PKA-downstream transcription factor CREB1. During EBOV infection, CREB1 is recruited into EBOV ribonucleoprotein complexes in viral inclusion bodies (VIBs) and employed for viral replication. AKIP1 depletion or PKA-CREB1 inhibition dramatically impairs EBOV replication. Meanwhile, the transcription of several coagulation-related genes, including THBD and SERPINB2, is substantially upregulated by VP35-dependent CREB1 activation, which may contribute to EBOV-related hemorrhage. The finding that EBOV VP35 hijacks the host PKA-CREB1 signal axis for viral replication and pathogenesis provides novel potential therapeutic approaches against EVD. Ebola virus virion protein 35 (VP35) is a cofactor of the viral RNA-dependent RNA polymerase, required for viral assembly and IFN antagonist. Here, Zhu et al. provide evidence that EBOV VP35 induces an AKIP1-mediated (human A kinase interacting protein) activation of the PKA-CREB1 signaling pathway and contributes to viral replication and pathogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Lin Zhu
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Ting Gao
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Yi Huang
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan, Hubei, 430020, China
| | - Jing Jin
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Di Wang
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Leike Zhang
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan, Hubei, 430020, China
| | - Yanwen Jin
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Ping Li
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Yong Hu
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Yan Wu
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan, Hubei, 430020, China
| | - Hainan Liu
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Qincai Dong
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Guangfei Wang
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Tong Zheng
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Caiwei Song
- Beijing Institute of Biotechnology, Beijing, 100039, China
| | - Yu Bai
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Xun Zhang
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Yaoning Liu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Weihong Yang
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Ke Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Gang Zou
- Insitut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lei Zhao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Gengfu Xiao
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan, Hubei, 430020, China.
| | - Xuan Liu
- Beijing Institute of Biotechnology, Beijing, 100039, China.
| | - Cheng Cao
- Beijing Institute of Biotechnology, Beijing, 100039, China.
| |
Collapse
|
20
|
Takaki W, Konishi H, Matsubara D, Shoda K, Arita T, Kataoka S, Shibamoto J, Furuke H, Takabatake K, Shimizu H, Komatsu S, Shiozaki A, Kubota T, Okamoto K, Otsuji E. Role of Extracellular High-Mobility Group Box-1 as a Therapeutic Target of Gastric Cancer. Int J Mol Sci 2022; 23:ijms23063264. [PMID: 35328684 PMCID: PMC8953630 DOI: 10.3390/ijms23063264] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
Background: High-mobility group box-1 (HMGB1) is involved in the tumorigenesis and metastasis of various cancers. The present study investigated the roles of extracellular HMGB1 in the progression of gastric cancer (GC) and the therapeutic effects of recombinant human soluble thrombomodulin (rTM) targeting HMGB1. Methods: The effects of extracellular HMGB1 and rTM on GC cells were assessed using proliferation and Transwell assays. Their effects on local tumor growth and metastasis were evaluated using subcutaneous tumor and liver metastasis mouse models, respectively. Plasma HMGB1 concentrations in GC patients were measured using ELISA. The relationships between plasma HMGB1 concentrations and the prognosis and clinicopathological factors of patients were also investigated. Results: GC proliferation, migration, and invasion abilities were promoted by increases in extracellular HMGB1 concentrations and alleviated by rTM. In the subcutaneous tumor model, local tumor growth was promoted by the addition of rhHMGB1 and alleviated by rTM. Similar changes occurred in the liver metastasis model. Recurrence-free survival (p < 0.01) and overall survival (p = 0.01) were significantly worse in patients with high plasma HMGB1 concentrations. Conclusion: Plasma HMGB1 concentrations are a prognostic marker in GC patients. Extracellular HMGB1 promotes cancer progression and has potential as a novel treatment target in GC cells for rTM.
Collapse
Affiliation(s)
- Wataru Takaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
- Correspondence: ; Tel.: +81-75-251-5527; Fax: +81-75-251-5522
| | - Daiki Matsubara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Katsutoshi Shoda
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Kofu 400-8510, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Satoshi Kataoka
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Jun Shibamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Hirotaka Furuke
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Kazuya Takabatake
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (W.T.); (D.M.); (K.S.); (T.A.); (S.K.); (J.S.); (H.F.); (K.T.); (H.S.); (S.K.); (A.S.); (T.K.); (K.O.); (E.O.)
| |
Collapse
|
21
|
Akwiwu E, Edem M, Akpotuzor J, Asemota E, Isong I. Glycated Haemogloin, Fasting Plasma Glucose, Plasminogen Activator Inhibitor Type-1, and Soluble Thrombomodulin Levels in Patients with Type 2 Diabetes Mellitus. Niger J Physiol Sci 2021; 36:159-164. [PMID: 35947738 DOI: 10.54548/njps.v36i2.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/16/2021] [Indexed: 06/15/2023]
Abstract
Diabetes mellitus has become increasingly prevalent over the years. The chronic hyperglycaemia of diabetes is associated with long-term damage, dysfunctions, and failure of different organs suggesting that the most effective tool to prevent complications is the effective control of hyperglycaemia itself. The study is set to determine the effect of glycemic control on plasminogen activator inhibitor type 1 (PAI-1), soluble thrombomodulin (STM) alongside fasting plasma glucose (FPG) and glycated hemoglobin (HbA1c) among type 2 diabetic subjects. One hundred diabetic subjects accessing care at the University of Calabar Teaching Hospital Calabar and 100 non -diabetics that served as controls were enrolled. Blood samples from participants were analyzed for FPG, HbA1c, PAI-1 and STM by standard methods. The result shows 74% of the diabetic to be females. Half of the diabetics were managed on only oral anti-diabetic drugs while the remaining half were either on insulin injection or a combination of oral and insulin injection. Poor glycemic control was observed in 56% of the studied subjects. The mean age of 54.69 ± 9.94 years for the diabetics was comparable to the age-matched controls (p=.097). Diabetics showed significantly higher FPG, HbA1c, PAI-1and STM (P=0.001) compared to control values. Correlations between STM, PAI 1 and glycated hemoglobin (figures 2 p=0.001, p =0.001) and STM, PAI-1 and FPG revealed significantly robust association (p=0.001, p=0.001). The study concludes that there is poor glycemic control among the treated diabetic subjects with PAI-1 and STM showing a very strong positive correlation with HbA1c than FPG.
Collapse
|
22
|
Mitaka C, Kawagoe I, Satoh D, Hayashida M. Effect of Recombinant Human Soluble Thrombomodulin on Coagulation-Related Variables in Patients With Sepsis-Induced Disseminated Intravascular Coagulation: A Retrospective Observational Study. Clin Appl Thromb Hemost 2021; 27:10760296211050356. [PMID: 34859680 PMCID: PMC8646186 DOI: 10.1177/10760296211050356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
To evaluate associations among coagulation-related variables, resolution of disseminated intravascular coagulation (DIC) and mortality, we retrospectively investigated 123 patients with sepsis-induced DIC treated with recombinant human soluble thrombomodulin (rTM). Changes in coagulation-related variables before and after treatment with rTM were examined. Further, associations between coagulation-related variables and DIC resolution were evaluated. The platelet count, prothrombin international normalized ratio (PT-INR), and fibrin/fibrinogen degradation products (FDP) significantly (p < .001) improved after rTM administration in survivors (n = 98), but not in nonsurvivors (n = 25). However, the DIC score significantly (p < .001) reduced in survivors and in nonsurvivors. Among coagulation-related variables examined before rTM, only PT-INR was significantly (p = .0395) lower in survivors than in nonsurvivors, and PT-INR before rTM was significantly (p = .0029) lower in patients attaining than not attaining DIC resolution (n = 87 and 36, respectively). The 28-day mortality was significantly lower in patients attaining than not attaining DIC resolution (11.5% vs 41.7%, p = .0001). In conclusion, the initiation of rTM administration before marked PT-INR elevation may be important to induce DIC resolution and thus to decrease mortality in patients with sepsis-induced DIC. Conversely, the treatment with rTM in patients with marked PT-INR elevation may be not so effective in achieving such goals.
Collapse
|
23
|
Berry J, Peaudecerf FJ, Masters NA, Neeves KB, Goldstein RE, Harper MT. An "occlusive thrombosis-on-a-chip" microfluidic device for investigating the effect of anti-thrombotic drugs. LAB ON A CHIP 2021; 21:4104-4117. [PMID: 34523623 PMCID: PMC8547327 DOI: 10.1039/d1lc00347j] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/05/2021] [Indexed: 05/03/2023]
Abstract
Cardiovascular disease remains one of the world's leading causes of death. Myocardial infarction (heart attack) is triggered by occlusion of coronary arteries by platelet-rich thrombi (clots). The development of new anti-platelet drugs to prevent myocardial infarction continues to be an active area of research and is dependent on accurately modelling the process of clot formation. Occlusive thrombi can be generated in vivo in a range of species, but these models are limited by variability and lack of relevance to human disease. Although in vitro models using human blood can overcome species-specific differences and improve translatability, many models do not generate occlusive thrombi. In those models that do achieve occlusion, time to occlusion is difficult to measure in an unbiased and objective manner. In this study we developed a simple and robust approach to determine occlusion time of a novel in vitro microfluidic assay. This highlighted the potential for occlusion to occur in thrombosis microfluidic devices through off-site coagulation, obscuring the effect of anti-platelet drugs. We therefore designed a novel occlusive thrombosis-on-a-chip microfluidic device that reliably generates occlusive thrombi at arterial shear rates by quenching downstream coagulation. We further validated our device and methods by using the approved anti-platelet drug, eptifibatide, recording a significant difference in the "time to occlude" in treated devices compared to control conditions. These results demonstrate that this device can be used to monitor the effect of antithrombotic drugs on time to occlude, and, for the first time, delivers this essential data in an unbiased and objective manner.
Collapse
Affiliation(s)
- Jess Berry
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.
| | - François J Peaudecerf
- Department of Civil, Environmental, and Geomatic Engineering, ETH Zürich, 8093 Zürich, Switzerland
| | - Nicole A Masters
- Department of Bioengineering, Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, Hemophilia and Thrombosis Center, University of Colorado Denver|Anschutz Medical Campus, Aurora, CO, USA
| | - Keith B Neeves
- Department of Bioengineering, Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, Hemophilia and Thrombosis Center, University of Colorado Denver|Anschutz Medical Campus, Aurora, CO, USA
| | - Raymond E Goldstein
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK.
| |
Collapse
|
24
|
Harmon AC, Noël A, Subramanian B, Perveen Z, Jennings MH, Chen YF, Penn AL, Legendre K, Paulsen DB, Varner KJ, Dugas TR. Inhalation of particulate matter containing free radicals leads to decreased vascular responsiveness associated with an altered pulmonary function. Am J Physiol Heart Circ Physiol 2021; 321:H667-H683. [PMID: 34415187 PMCID: PMC8794232 DOI: 10.1152/ajpheart.00725.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022]
Abstract
Airborne particulate matter (PM) is associated with an increased risk for cardiovascular diseases. Although the goal of thermal remediation is to eliminate organic wastes through combustion, when incomplete combustion occurs, organics chemisorb to transition metals to generate PM-containing environmentally persistent free radicals (EPFRs). Similar EPFR species have been detected in PM found in diesel and gasoline exhaust, woodsmoke, and urban air. Prior in vivo studies demonstrated that EPFRs reduce cardiac function secondary to elevations in pulmonary arterial pressures. In vitro studies showed that EPFRs increase ROS and cytokines in pulmonary epithelial cells. We thus hypothesized that EPFR inhalation would promote lung inflammation and oxidative stress, leading to systemic inflammation, vascular endothelial injury, and a decline in vascular function. Mice were exposed to EPFRs for either 4 h or for 4 h/day for 10 days and lung and vascular function were assessed. After a 4-h exposure, plasma nitric oxide (NO) was reduced while endothelin-1 (ET-1) was increased, however lung function was not altered. After 10 day, plasma NO and ET-1 levels were again altered and lung tidal volume was reduced. These time course studies suggested the vasculature may be an early target of injury. To test this hypothesis, an intermediate time point of 3 days was selected. Though the mice exhibited no marked inflammation in either the lung or the blood, we did note significantly reduced endothelial function concurrent with a reduction in lung tidal volume and an elevation in annexin V protein levels in the lung. Although vascular dysfunction was not dependent upon inflammation, it may be associated with an injury at the air-blood interface. Gene expression analysis suggested roles for oxidative stress and aryl hydrocarbon receptor (Ahr) signaling. Studies probing the relationship between pulmonary oxidative stress and AhR signaling at the air-blood interface with vascular dysfunction seem warranted.NEW & NOTEWORTHY Particulate matter (PM) resulting from the combustion of organic matter is known to contribute to cardiopulmonary disease. Despite hypotheses that cardiovascular dysfunction occurring after PM exposures is secondary to lung or systemic inflammation, these studies investigating exposures to PM-containing environmentally persistent free radicals (EPFRs) demonstrate that cardiovascular dysfunction precedes pulmonary inflammation. The cardiopulmonary health consequences of EPFRs have yet to be thoroughly evaluated, especially in healthy, adult mice. Our data suggest the vasculature as a direct target of PM exposure, and our studies aimed to elucidate the mechanisms contributing to EPFR-induced vascular dysfunction.
Collapse
Affiliation(s)
- Ashlyn C Harmon
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | | | - Zakia Perveen
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Merilyn H Jennings
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Yi-Fan Chen
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Arthur L Penn
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Kelsey Legendre
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Daniel B Paulsen
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Kurt J Varner
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Tammy R Dugas
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| |
Collapse
|
25
|
Hambley BC, Tomuleasa C, Ghiaur G. Coagulopathy in Acute Promyelocytic Leukemia: Can We Go Beyond Supportive Care? Front Med (Lausanne) 2021; 8:722614. [PMID: 34485349 PMCID: PMC8415964 DOI: 10.3389/fmed.2021.722614] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/26/2021] [Indexed: 11/24/2022] Open
Abstract
Acute promyelocytic leukemia (APL) is characterized by frequent complications due to a distinct coagulopathy. While advances in treatments have improved long-term survival, hemorrhagic and thrombotic complications remain the most common causes of death and morbidity. Improved understanding of the mechanisms of the coagulopathy associated with APL may lead to therapeutic interventions to mitigate the risk of hemorrhage and thrombosis.
Collapse
Affiliation(s)
- Bryan C Hambley
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
26
|
Che Mohd Nassir CMN, Hashim S, Wong KK, Abdul Halim S, Idris NS, Jayabalan N, Guo D, Mustapha M. COVID-19 Infection and Circulating Microparticles-Reviewing Evidence as Microthrombogenic Risk Factor for Cerebral Small Vessel Disease. Mol Neurobiol 2021; 58:4188-4215. [PMID: 34176095 PMCID: PMC8235918 DOI: 10.1007/s12035-021-02457-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/16/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) due to novel coronavirus disease 2019 (COVID-19) has affected the global society in numerous unprecedented ways, with considerable morbidity and mortality. Both direct and indirect consequences from COVID-19 infection are recognized to give rise to cardio- and cerebrovascular complications. Despite current limited knowledge on COVID-19 pathogenesis, inflammation, endothelial dysfunction, and coagulopathy appear to play critical roles in COVID-19-associated cerebrovascular disease (CVD). One of the major subtypes of CVD is cerebral small vessel disease (CSVD) which represents a spectrum of pathological processes of various etiologies affecting the brain microcirculation that can trigger subsequent neuroinflammation and neurodegeneration. Prevalent with aging, CSVD is a recognized risk factor for stroke, vascular dementia, and Alzheimer's disease. In the background of COVID-19 infection, the heightened cellular activations from inflammations and oxidative stress may result in elevated levels of microthrombogenic extracellular-derived circulating microparticles (MPs). Consequently, MPs could act as pro-coagulant risk factor that may serve as microthrombi for the vulnerable microcirculation in the brain leading to CSVD manifestations. This review aims to appraise the accumulating body of evidence on the plausible impact of COVID-19 infection on the formation of microthrombogenic MPs that could lead to microthrombosis in CSVD manifestations, including occult CSVD which may last well beyond the pandemic era.
Collapse
Affiliation(s)
- Che Mohd Nasril Che Mohd Nassir
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Sabarisah Hashim
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Kah Keng Wong
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Sanihah Abdul Halim
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nur Suhaila Idris
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nanthini Jayabalan
- Translational Neuroscience Lab, UQ Centre for Clinical Research, the University of Queensland, Herston, Brisbane, 4029, Australia
| | - Dazhi Guo
- Department of Hyperbaric Oxygen, The Sixth Medical Center of PLA General Hospital, 6 Fucheng Rd, Beijing, 100048, China
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
27
|
Li Q, Yang W, Zhao K, Sun X, Bao L. Thrombomodulin gene polymorphism and the occurrence and prognostic value of sepsis acute kidney injury. Medicine (Baltimore) 2021; 100:e26293. [PMID: 34190147 PMCID: PMC8257907 DOI: 10.1097/md.0000000000026293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 04/20/2021] [Accepted: 05/19/2021] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT To investigate the relationship between thrombomodulin (THBD) gene single nucleotide polymorphisms (SNPs) and susceptibility to sepsis and the occurrence and prognosis of acute kidney injury (AKI) in sepsis patients.The genotypes of THBD gene rs1962, rs3176123, and rs1042580 in 178 sepsis patients with AKI, 243 sepsis patients without AKI (No AKI), and 103 healthy controls were analyzed by direct sequencing. Enzyme-linked immunosorbent assay (ELISA) was used to detect the plasma THBD protein levels. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic value of plasma THBD levels in sepsis, AKI, and death of sepsis patients.The C allele carriers of THBD gene rs1962 were more likely to develop AKI and sepsis than the T allele carriers (OR = 1.61, 95% CI: 1.18-2.19, P < .01; OR = 2.16, 95% CI: 1.42-3.29, P < .01). The rs3176123 G allele was associated with an increased risk of AKI in sepsis patients (OR = 1.41, 95% CI: 1.06-1.88, P = .02), the G allele had a significant association with a higher risk of sepsis susceptibility (OR = 1.91, 95% CI: 1.33-2.75, P < .01). Sepsis patients of rs1042580 C allele had a lower risk of AKI than those of T allele (OR = 0.58, 95% CI: 0.37-0.91, P = .02), the C allele was related to a reduced risk of sepsis susceptibility (OR = 0.38, 95% CI: 0.26-0.55, P < .01). The THBD gene rs1962, rs3176123, and rs1042580 TGT haplotype was linked to higher risk of AKI in patients with sepsis (OR = 1.96, 95%CI: 1.14-3.38, P = .02). Sepsis patients with the THBD gene rs1962 TC + CC genotype had a higher risk of death than those with TT genotype (OR = 10.93, 95%CI: 5.05-26.96, P < .01), but there was no significant difference in the risk of death in sepsis patients with different genotypes at rs3176123 and rs1042580 (P > .05).The THBD gene rs1962, rs3176123, and rs1042580 SNPs are significantly associated with sepsis susceptibility and the risk of AKI. The rs1962 SNP is related to the risk of death in sepsis patients.
Collapse
Affiliation(s)
- Qin Li
- Department of Nephrology Ward, Zibo Central Hospital, Zhangdian, Zibo, Shandong
| | - Wenjuan Yang
- Department of Nephrology Ward, Zibo Central Hospital, Zhangdian, Zibo, Shandong
| | - Keming Zhao
- Department of Nephrology Ward, Zibo Central Hospital, Zhangdian, Zibo, Shandong
| | - Xifeng Sun
- Department of Nephrology Ward, Zibo Central Hospital, Zhangdian, Zibo, Shandong
| | - Liuqian Bao
- Department of Emergency Medicine Department, People's Hospital of Tiantai County, Tiantai County, Taizhou City, Zhejiang, China
| |
Collapse
|
28
|
Hara H, Iwase H, Nguyen H, Miyagawa Y, Kuravi K, Foote JB, Eyestone W, Phelps C, Ayares D, Cooper DKC. Stable expression of the human thrombomodulin transgene in pig endothelial cells is associated with a reduction in the inflammatory response. Cytokine 2021; 148:155580. [PMID: 34099346 DOI: 10.1016/j.cyto.2021.155580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND Xenotransplantation is associated with an inflammatory response. The proinflammatory cytokine, TNF-α, downregulates the expression of thrombomodulin (TBM), and induces coagulation dysfunction. Although human (h) TBM-transgenic pigs (p) have been developed to reduce coagulation dysfunction, the effect of TNF-α on the expression of hTBM and its functional activity has not been fully investigated. The aims of this study were to investigate (i) whether the expression of hTBM on pig (p) cells is down-regulated during TNF-α stimulation, and (ii) whether cells from hTBM pigs regulate the inflammatory response. METHODS TNF-α-producing T, B, and natural killer cells in blood from baboons with pig heart or kidney xenografts were investigated by flow cytometry. TNF-α staining in the grafts was detected by immunohistochemistry. Aortic endothelial cells (AECs) from GTKO/CD46 and GTKO/CD46/hTBM pigs were stimulated by hTNF-α, and the expression of the inflammatory/coagulation regulatory protein, TBM, was investigated. RESULTS After pig organ xenotransplantation, there was a trend to increases in TNF-α-producing T and natural killer cells in the blood of baboons. In vitro observations demonstrated that after hTNF-α stimulation, there was a significant reduction in the expression of endogenous pTBM on pAECs, and a significant increase in the expression of inflammatory molecules. Blocking of NF-κB signaling significantly up-regulated pTBM expression, and suppressed the inflammatory response induced by hTNF-α in pAECs. Whereas the expression of pTBM mRNA was significantly reduced by hTNF-α stimulation, hTBM expression on the GTKO/CD46/hTBM pAECs was not affected. Furthermore, after hTNF-α stimulation, there was significant suppression of expression of inflammatory molecules on GTKO/CD46/hTBM pAECs compared to GTKO/CD46 pAECs. CONCLUSIONS The stable expression of hTBM in pig cells may locally regulate the inflammatory response. This will help suppress the inflammatory response and prevent coagulation dysregulation after xenotransplantation.
Collapse
Affiliation(s)
- Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huy Nguyen
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuko Miyagawa
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
29
|
Meng S, Kang K, Fei D, Yang S, Gu Q, Pan S, Zhao M. Preliminary study of microparticle coagulation properties in septic patients with disseminated intravascular coagulation. J Int Med Res 2021; 49:3000605211014094. [PMID: 34034547 PMCID: PMC8161883 DOI: 10.1177/03000605211014094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background Sepsis typically results in enhanced coagulation system activation and microthrombus formation. Microparticle (MP) production promotes coagulation and enhances pro-coagulation. This study investigated how circulating MP levels and tissue factor-bearing MP (TF+-MP) activity caused coagulation in patients with septic disseminated intravascular coagulation (DIC). Methods Thirty patients with septic DIC and 30 healthy controls were studied from December 2017 to March 2019. Patient blood samples were collected at enrolment (day 1) and on days 3 and 5; DIC scores and Sequential Organ Failure Assessment (SOFA) scores were recorded. TF+-MP activity was measured using TF-dependent factor Xa generation experiments. Circulating MP concentrations were determined by MP capture assay. Clotting factor activity, antithrombin level, soluble thrombomodulin, and serum tissue factor pathway inhibitor (TFPI) concentrations were measured. Results Patients with septic DIC had lower circulating MP levels than healthy control patients. Circulating MP levels in patients with septic DIC were positively correlated with DIC scores and negatively correlated with coagulation factors, but TF+-MP activity did not correlate with clotting factor levels and TFPI. Conclusions In patients with septic DIC, circulating MP levels are important in promoting coagulation activation and increasing clotting factor consumption. TF+-MP activity may not be the main form of active TF.
Collapse
Affiliation(s)
- Shishuai Meng
- Department of Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Kai Kang
- Department of Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongsheng Fei
- Department of Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Songlin Yang
- Department of Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Quankuan Gu
- Department of Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - ShangHa Pan
- The Key Hepatosplenic Surgery Laboratory, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Mingyan Zhao
- Department of Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
30
|
Li Y, Wan D, Luo X, Song T, Wang Y, Yu Q, Jiang L, Liao R, Zhao W, Su B. Circulating Histones in Sepsis: Potential Outcome Predictors and Therapeutic Targets. Front Immunol 2021; 12:650184. [PMID: 33868288 PMCID: PMC8044749 DOI: 10.3389/fimmu.2021.650184] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/11/2021] [Indexed: 02/05/2023] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection and is associated with high morbidity and mortality. Circulating histones (CHs), a group of damage-associated molecular pattern molecules mainly derived from neutrophil extracellular traps, play a crucial role in sepsis by mediating inflammation response, organ injury and death through Toll-like receptors or inflammasome pathways. Herein, we first elucidate the molecular mechanisms of histone-induced inflammation amplification, endothelium injury and cascade coagulation activation, and discuss the close correlation between elevated level of CHs and disease severity as well as mortality in patients with sepsis. Furthermore, current state-of-the-art on anti-histone therapy with antibodies, histone-binding proteins (namely recombinant thrombomodulin and activated protein C), and heparin is summarized to propose promising approaches for sepsis treatment.
Collapse
Affiliation(s)
- Yupei Li
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China.,Department of Emergency Medicine of West China Hospital, Disaster Medical Center, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Dingyuan Wan
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xinyao Luo
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Song
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Yiran Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Qiao Yu
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China.,Department of Emergency Medicine of West China Hospital, Disaster Medical Center, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Luojia Jiang
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Ruoxi Liao
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China.,Department of Emergency Medicine of West China Hospital, Disaster Medical Center, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Wang Z, Jiang L, Yan H, Xu Z, Luo P. Adverse events associated with nilotinib in chronic myeloid leukemia: mechanisms and management strategies. Expert Rev Clin Pharmacol 2021; 14:445-456. [PMID: 33618586 DOI: 10.1080/17512433.2021.1894129] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Introduction: Nilotinib is a second-generation tyrosine kinase inhibitor (TKI) targeting BCR/ABL, which is used for the first-line treatment of newly diagnosed chronic myeloid leukemia (CML) patients and the second-line treatment of most CML patients who are resistant or intolerant to prior therapy that includes imatinib. In addition to common adverse reactions, long-term use of nilotinib shows some toxicities that are different from those of occurring during other BCR/ABL TKI treatments, such as cardiovascular toxicity. It is life-threatening, which would affect not only the choice of initial treatment of CML patients but also the safety of long-term medication.Areas covered: Through searching literature and reports from PubMed and clinical trials, here we review a profile of the adverse effects induced by nilotinib. We also discuss the potential molecular toxicological mechanisms and clinical management, which may provide strategies to prevent or intervene the toxicity associated with nilotinib.Expert opinion: Severe adverse effects associated with nilotinib limit its long-term clinical application. However, the exact mechanisms underlying these toxicities remain unclear. Future research should focus on the developing strategies to reduce the toxicities of nilotinib as well as to avoid similar toxicity in the development of new drugs.
Collapse
Affiliation(s)
- Zeng Wang
- Department of Colorectal Pharmacy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Liyu Jiang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Kanou S, Miyashita T, Yamamoto Y, Takada S, Nakura M, Okazaki M, Ohbatake Y, Nakanuma S, Makino I, Tajima H, Takamura H, Fushida S, Ohta T. Prophylactic Effect of Recombinant Human Soluble Thrombomodulin for Hepatic Sinusoidal Obstruction Syndrome Model Mice. In Vivo 2021; 34:1037-1045. [PMID: 32354890 DOI: 10.21873/invivo.11873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023]
Abstract
AIM The present study aimed to examine the effects of prophylactic administration of recombinant human soluble thrombomodulin (rTM) for the prevention of sinusoidal obstruction syndrome (SOS). MATERIALS AND METHODS Crl:CD1 mice were allocated to the rTM, placebo, and control groups. The rTM group received an intraperitoneal administration of rTM, with intraperitoneal administration of monocrotaline (MCT) 1 h later. The placebo group received PBS instead of rTM, and the control group received PBS instead of rTM and MCT. Mice were sacrificed 48 h after MCT administration, and blood and liver tissues were evaluated. Immunostaining was performed using anti-CD42b and anti-SE-1 antibodies, and AZAN staining. Levels of plasminogen activator inhibitor (PAI-1) and endothelial nitric oxide synthase (eNOS) in whole liver tissues were estimated using RT-PCR. RESULTS Hematoxylin-eosin staining showed that SOS-related findings were markedly attenuated in the rTM group compared to the placebo group. CD42b immunostaining showed the presence of extravasated platelet activation (EPA) in the Disse space in the placebo group, but this was less noticeable in the rTM group. PAI-1 levels were significantly lower in the rTM group than in the placebo group in RT-PCR. However, eNOS levels were significantly higher in the rTM group than in the placebo group. CONCLUSION Administration of rTM may prevent SOS by protecting sinusoidal endothelial cells.
Collapse
Affiliation(s)
- Shunsuke Kanou
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Satoshi Takada
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Makoto Nakura
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Mitsuyoshi Okazaki
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Yoshinao Ohbatake
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Shinichi Nakanuma
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|
33
|
Mihalko EP, Sandry M, Mininni N, Nellenbach K, Deal H, Daniele M, Ghadimi K, Levy JH, Brown AC. Fibrin-modulating nanogels for treatment of disseminated intravascular coagulation. Blood Adv 2021; 5:613-627. [PMID: 33560377 PMCID: PMC7876887 DOI: 10.1182/bloodadvances.2020003046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Disseminated intravascular coagulation (DIC) is a pathological coagulopathy associated with infection that increases mortality. In DIC, excessive thrombin generation causes symptoms from formation of microthrombi to multiorgan failure; bleeding risks can also be a concern because of clotting factor consumption. Different clinical events lead to DIC, including sepsis, trauma, and shock. Treatments for thrombotic episodes or bleeding presentation in DIC oppose each other, thus creating therapeutic dilemmas in management. The objective of this study was to develop fibrin-specific core-shell nanogels (FSNs) loaded with tissue-type plasminogen activator (tPA) to treat the microcirculatory complications of DIC, which would facilitate targeted clot dissolution to manage microthrombi and the potential consumptive coagulopathy that causes bleeding. FSNs enhance formation of actively polymerizing clots by crosslinking fibrin fibers, but they can also target preexisting microthrombi and, when loaded with tPA, facilitate targeted delivery to lyse the microthrombi. We hypothesized that this dual action would simultaneously address bleeding and microthrombi with DIC to improve outcomes. In vivo, tPA-FSNs decreased the presentation of multiorgan microthrombi, recovered platelet counts, and improved bleeding outcomes in a DIC rodent model. When incorporated with human DIC patient plasma, tPA-FSNs restored clot structure and clot growth under flow. Together, these data demonstrate that a fibrinolytic agent loaded into fibrin-targeting nanogels could improve DIC outcomes.
Collapse
Affiliation(s)
- Emily P Mihalko
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
- Comparative Medicine Institute and
| | - Megan Sandry
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
| | - Nicholas Mininni
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
| | - Kimberly Nellenbach
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
- Comparative Medicine Institute and
| | - Halston Deal
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
- Comparative Medicine Institute and
| | - Michael Daniele
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
- Comparative Medicine Institute and
- Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, NC; and
| | - Kamrouz Ghadimi
- Department of Anesthesiology and Critical Care, Duke University School of Medicine, Durham, NC
| | - Jerrold H Levy
- Department of Anesthesiology and Critical Care, Duke University School of Medicine, Durham, NC
| | - Ashley C Brown
- Joint Department of Biomedical Engineering of University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC
- Comparative Medicine Institute and
| |
Collapse
|
34
|
Kawasoe J, Uchida Y, Miyauchi T, Kadono K, Hirao H, Saga K, Watanabe T, Ueda S, Terajima H, Uemoto S. The lectin-like domain of thrombomodulin is a drug candidate for both prophylaxis and treatment of liver ischemia and reperfusion injury in mice. Am J Transplant 2021; 21:540-551. [PMID: 32805077 PMCID: PMC7891328 DOI: 10.1111/ajt.16269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/13/2020] [Accepted: 08/03/2020] [Indexed: 01/25/2023]
Abstract
Ischemia and reperfusion injury (IRI) can occur in any tissue or organ. With respect to liver transplantation, the liver grafts from donors by definition experience transient ischemia and subsequent blood reflow. IRI is a problem not only in organ transplantation but also in cases of thrombosis or circulatory disorders such as mesenteric ischemia, myocardial, or cerebral infarction. We have reported that recombinant human soluble thrombomodulin (rTM), which is currently used in Japan to treat disseminated intravascular coagulation (DIC), has a protective effect and suppresses liver IRI in mice. However, rTM may not be fully safe to use in humans because of its inherent anticoagulant activity. In the present study, we used a mouse liver IRI model to explore the possibility that the isolated lectin-like domain of rTM (rTMD1), which has no anticoagulant activity, could be effective as a therapeutic modality for IRI. Our results indicated that rTMD1 could suppress ischemia and reperfusion-induced liver damage in a dose-dependent manner without concern of associated hemorrhage. Surprisingly, rTMD1 suppressed the liver damage even after IR insult had occurred. Taken together, we conclude that rTMD1 may be a candidate drug for prevention of and therapy for human liver IRI without the possible risk of hemorrhage.
Collapse
Affiliation(s)
- Junya Kawasoe
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan,Department of Gastroenterological Surgery and OncologyThe Tazuke Kofukai Medical Research Institute, Kitano HospitalOsakaJapan
| | - Yoichiro Uchida
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan,Department of Gastroenterological Surgery and OncologyThe Tazuke Kofukai Medical Research Institute, Kitano HospitalOsakaJapan
| | - Tomoyuki Miyauchi
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan,Department of Gastroenterological Surgery and OncologyThe Tazuke Kofukai Medical Research Institute, Kitano HospitalOsakaJapan
| | - Kentaro Kadono
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hirofumi Hirao
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kenichi Saga
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan,Department of Gastroenterological Surgery and OncologyThe Tazuke Kofukai Medical Research Institute, Kitano HospitalOsakaJapan
| | - Takeshi Watanabe
- Division of Immunology, Institute for Frontier Life and Medical SciencesKyoto UniversityKyotoJapan
| | - Shugo Ueda
- Department of Gastroenterological Surgery and OncologyThe Tazuke Kofukai Medical Research Institute, Kitano HospitalOsakaJapan
| | - Hiroaki Terajima
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan,Department of Gastroenterological Surgery and OncologyThe Tazuke Kofukai Medical Research Institute, Kitano HospitalOsakaJapan
| | - Shinji Uemoto
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
35
|
Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, He Y, Chopra A, Chieng HC, Tiwari A, Judson MA, Paulson B, Brademan DR, Zhu Y, Serrano LR, Linke V, Drake LA, Adam AP, Schwartz BS, Singer HA, Swanson S, Mosher DF, Stewart R, Coon JJ, Jaitovich A. Large-Scale Multi-omic Analysis of COVID-19 Severity. Cell Syst 2021; 12:23-40.e7. [PMID: 33096026 PMCID: PMC7543711 DOI: 10.1016/j.cels.2020.10.003] [Citation(s) in RCA: 397] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/24/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
We performed RNA-seq and high-resolution mass spectrometry on 128 blood samples from COVID-19-positive and COVID-19-negative patients with diverse disease severities and outcomes. Quantified transcripts, proteins, metabolites, and lipids were associated with clinical outcomes in a curated relational database, uniquely enabling systems analysis and cross-ome correlations to molecules and patient prognoses. We mapped 219 molecular features with high significance to COVID-19 status and severity, many of which were involved in complement activation, dysregulated lipid transport, and neutrophil activation. We identified sets of covarying molecules, e.g., protein gelsolin and metabolite citrate or plasmalogens and apolipoproteins, offering pathophysiological insights and therapeutic suggestions. The observed dysregulation of platelet function, blood coagulation, acute phase response, and endotheliopathy further illuminated the unique COVID-19 phenotype. We present a web-based tool (covid-omics.app) enabling interactive exploration of our compendium and illustrate its utility through a machine learning approach for prediction of COVID-19 severity.
Collapse
Affiliation(s)
- Katherine A Overmyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Morgridge Institute for Research, Madison, WI 53562, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ian J Miller
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Joseph Balnis
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | | | - Trenton M Peters-Clarke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Jesse G Meyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Qiuwen Quan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Laura K Muehlbauer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Edna A Trujillo
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yuchen He
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Amit Chopra
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Hau C Chieng
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Anupama Tiwari
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Division of Sleep Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Marc A Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Brett Paulson
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Dain R Brademan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yunyun Zhu
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lia R Serrano
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Vanessa Linke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lisa A Drake
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; Department of Ophthalmology, Albany Medical College, Albany, NY 12208, USA
| | | | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Scott Swanson
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Deane F Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Joshua J Coon
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Morgridge Institute for Research, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA.
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
36
|
Şahin A, Özer N. The effect of activated protein C in the experimental disseminated intravascular coagulation model formed by lipopolysaccharide infusion. Acta Cir Bras 2020; 35:e351102. [PMID: 33331452 PMCID: PMC7748076 DOI: 10.1590/acb351102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/04/2020] [Indexed: 03/17/2023] Open
Abstract
Purpose: In this experimental study, activated protein C (APC), which has anticoagulant, antithrombotic, profibrinolytic, anti-inflammatory and antiapoptotic properties, was used to prevent coagulopathy in a disseminated intravascular coagulation (DIC) model formatted with lipopolysaccharide (LPS) infusion. Methods: Twenty-five Wistar albino rats weighting 280 – 320 g each were used. They were randomly divided into three groups: sham, control and study groups. To sham group (n = 5), only normal saline was infused. To control (n = 10) and study groups (n = 10), 30 mg/kg LPS was infused for 4 h from femoral vein. After LPS infusion, 100 µg/kg recombinant APC was given during 4 h in study group. Eight hours later, blood samples were taken from abdominal aorta and the animals sacrificed. From these samples, platelet, prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen and D-dimer levels were studied. Results: Platelet counts and fibrinogen levels were significantly lower in control and study groups than sham group (p < 0.05). The PT, aPTT and D-dimer levels were significantly higher in control and study groups than in sham group (p < 0.05). When comparing control and study groups, platelet counts were not statistically different (p = 0.36). However, the difference of the fibrinogen levels was significant between these groups (p = 0.0001). While PT and aPTT were longer in the study group compared to the control group (p < 0.05), D-dimer levels were lower in the study group than in control (p = 0.0001). Conclusion: Use of APC can prevent hypercoagulation and consumption coagulopathy in the DIC as a result of correcting hematological parameters other than prolongation of coagulation time.
Collapse
|
37
|
The effect of recombinant human soluble thrombomodulin on renal function and mortality in septic disseminated intravascular coagulation patients with acute kidney injury: a retrospective study. J Intensive Care 2020; 8:94. [PMID: 33308326 PMCID: PMC7729679 DOI: 10.1186/s40560-020-00512-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/04/2020] [Indexed: 12/25/2022] Open
Abstract
Background Clinical evidence showing the effectiveness of recombinant human soluble thrombomodulin (rhTM) for treating sepsis-induced disseminated intravascular coagulation (DIC) and organ dysfunction (particularly renal injury) is limited because of differences in the inclusion criteria and disease severity among patients. This study aimed to assess the association between rhTM and outcomes in septic DIC patients with acute kidney injury (AKI). Methods This retrospective observational study analyzed the data of patients who were admitted to the intensive care unit (ICU) of a single center between January 2012 and December 2018, and diagnosed with sepsis-induced DIC and AKI. Data were extracted as follows: patients’ characteristics; DIC score, as calculated by the Japanese Association for Acute Medicine and the International Society of Thrombosis and Hemostasis criteria; serum creatinine levels; and ICU and 28-day mortality rates. The primary outcome was the dependence on renal replacement therapy (RRT) at ICU discharge. The propensity score (PS) was calculated using the following variables: age, sex, septic shock at admission, DIC score, and KDIGO classification. Subsequently, logistic regression analysis was performed using the PS to evaluate the outcome. Results In total, 97 patients were included in this study. Of these, 52 (53.6%) patients had received rhTM. The dependence on RRT at ICU discharge was significantly lower in the rhTM than in the non-rhTM group (odds ratio [OR], 0.43; 95% confidence interval [CI], 0.19–0.97; P = 0.043). The serum creatinine levels at ICU discharge (OR, 0.31; 95% CI, 0.13–0.72; P = 0.007) and hospital discharge (OR, 0.25; 95% CI, 0.11–0.60; P = 0.002, respectively), and the 28-day mortality rate (OR, 0.40; 95% CI, 0.17–0.93; P = 0.033) were significantly lower in the rhTM than in the non-rhTM group. Moreover, the Kaplan–Meier survival curve revealed significantly lower mortality rates in the rhTM than in the non-rhTM group (P = 0.009). No significant differences in the DIC score and AKI severity were observed between the groups. Conclusions Among sepsis-induced DIC patients with AKI, rhTM administration was associated with lower dependence on RRT at ICU discharge, improvement in renal function, and lower 28-day mortality rate.
Collapse
|
38
|
Abstract
Endothelial cells (ECs) are vascular, nonconventional immune cells that play a major role in the systemic response after bacterial infection to limit its dissemination. Triggered by exposure to pathogens, microbial toxins, or endogenous danger signals, EC responses are polymorphous, heterogeneous, and multifaceted. During sepsis, ECs shift toward a proapoptotic, proinflammatory, proadhesive, and procoagulant phenotype. In addition, glycocalyx damage and vascular tone dysfunction impair microcirculatory blood flow, leading to organ injury and, potentially, life-threatening organ failure. This review aims to cover the current understanding of the EC adaptive or maladaptive response to acute inflammation or bacterial infection based on compelling recent basic research and therapeutic clinical trials targeting microvascular and endothelial alterations during septic shock.
Collapse
Affiliation(s)
- Jérémie Joffre
- Medical Intensive Care Unit, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France.,Department of Anesthesia and Perioperative Care, University of California San Francisco School of Medicine, San Francisco, California
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California San Francisco School of Medicine, San Francisco, California
| | - Can Ince
- Department of Intensive Care Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, the Netherlands; and
| | - Hafid Ait-Oufella
- Medical Intensive Care Unit, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France.,INSERM U970, Cardiovascular Research Center, Université de Paris, Paris, France
| |
Collapse
|
39
|
Sriram K, Insel PA. Inflammation and thrombosis in COVID-19 pathophysiology: proteinase-activated and purinergic receptors as drivers and candidate therapeutic targets. Physiol Rev 2020; 101:545-567. [PMID: 33124941 PMCID: PMC8238137 DOI: 10.1152/physrev.00035.2020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Paul A Insel
- Department of Pharmacology and Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
40
|
Katz JM, Libman RB, Wang JJ, Filippi CG, Sanelli P, Zlochower A, Gribko M, Pacia SV, Kuzniecky RI, Najjar S, Azhar S. COVID-19 Severity and Stroke: Correlation of Imaging and Laboratory Markers. AJNR Am J Neuroradiol 2020; 42:257-261. [PMID: 33122216 DOI: 10.3174/ajnr.a6920] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE Coronavirus disease 2019 (COVID-19) appears to be an independent risk factor for stroke. We hypothesize that patients who develop stroke while hospitalized for severe COVID-19 will have higher inflammatory markers and distinct stroke imaging patterns compared with patients positive for COVID-19 with out-of-hospital stroke onset and milder or no COVID-19 symptoms. MATERIALS AND METHODS This is a retrospective case series of patients positive for COVID-19 on polymerase chain reaction testing with imaging-confirmed stroke treated within a large health care network in New York City and Long Island between March 14 and April 26, 2020. Clinical and laboratory data collected retrospectively included complete blood counts and creatinine, alanine aminotransferase, lactate dehydrogenase, C-reactive protein, ferritin, and D-dimer levels. All CT and MR imaging studies were independently reviewed by 2 neuroradiologists who recorded stroke subtype and patterns of infarction and intracranial hemorrhage. RESULTS Compared with patients with COVID-19 with outside-of-hospital stroke onset and milder or no COVID-19 symptoms (n = 45, 52.3%), patients with stroke already hospitalized for severe COVID-19 (n = 41, 47.7%) had significantly more frequent infarctions (95.1% versus 73.3%, P = .006), with multivascular distributions (56.4% versus 33.3%, P = .022) and associated hemorrhage (31.7% versus 4.4%, P = .001). Patients with stroke admitted with more severe COVID-19 had significantly higher C-reactive protein and ferritin levels, elevated D-dimer levels, and more frequent lymphopenia and renal and hepatic injury (all, P < .003). CONCLUSIONS Patients with stroke hospitalized with severe COVID-19 are characterized by higher inflammatory, coagulopathy, and tissue-damage biomarkers, supporting proposed pathogenic mechanisms of hyperinflammation activating a prothrombotic state. Cautious balancing of thrombosis and the risk of hemorrhagic transformation is warranted when considering anticoagulation.
Collapse
Affiliation(s)
- J M Katz
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| | - R B Libman
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| | - J J Wang
- Feinstein Institute for Medical Research at Northwell Health (J.J.W.), Manhasset, New York
| | - C G Filippi
- Radiology (C.G.F., P.S., A.Z.), Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - P Sanelli
- Radiology (C.G.F., P.S., A.Z.), Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - A Zlochower
- Radiology (C.G.F., P.S., A.Z.), Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - M Gribko
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| | - S V Pacia
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| | - R I Kuzniecky
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| | - S Najjar
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| | - S Azhar
- From the Department of Neurology (J.M.K., R.B.L., M.G., S.V.P., R.I.K., S.N., S.A.)
| |
Collapse
|
41
|
Ikezoe T. Advances in the diagnosis and treatment of disseminated intravascular coagulation in haematological malignancies. Int J Hematol 2020; 113:34-44. [PMID: 32902759 DOI: 10.1007/s12185-020-02992-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/22/2020] [Accepted: 08/28/2020] [Indexed: 11/26/2022]
Abstract
Haematological malignancies, including acute leukaemia and non-Hodgkin lymphoma, are one of the underlying diseases that frequently cause disseminated intravascular coagulation (DIC), an acquired thrombotic disorder. Concomitant DIC is associated with the severity of the underlying disease and poor prognosis. The Japanese Society on Thrombosis and Hemostasis released the new DIC diagnostic criteria in 2017. This criteria include coagulation markers such as soluble fibrin and the thrombin-antithrombin complex to more accurately evaluate the hypercoagulable state in patients. Among several groups of anticoagulants available, recombinant human soluble thrombomodulin is most frequently used to treat DIC caused by haematological malignancies in Japan. DIC is remitted in parallel with the improvement of the underlying haematological diseases; thus, there is room for debate regarding whether the treatment of DIC would improve the prognosis of patients. Haematopoietic stem cell transplantation as well as the recently introduced chimeric antigen receptor (CAR)-T-cell therapy are innovative therapies to produce a cure in a subset of patients with haematological malignancies. However, coagulopathy frequently occurs after these therapies, which limits the success of the treatment. For example, DIC is noted in approximately 50% of patients after CAT-T-cell therapy in conjunction with cytokine release syndrome. Hematopoietic stem cell transplantation (HSCT) causes endotheliitis, which triggers coagulopathy and the development of potentially lethal complications, such as sinusoidal obstruction syndrome/veno-occlusive disease and transplant-associated thrombotic microangiopathy. This review article describes the pathogenesis, clinical manifestation, diagnosis, and treatment of DIC caused by haematological malignancies, CAR-T-cell therapy, and HSCT.
Collapse
Affiliation(s)
- Takayuki Ikezoe
- Department of Haematology, Fukushima Medical University, Fukushima, 960-1295, Japan.
| |
Collapse
|
42
|
Ahmed S, Zimba O, Gasparyan AY. Thrombosis in Coronavirus disease 2019 (COVID-19) through the prism of Virchow's triad. Clin Rheumatol 2020; 39:2529-2543. [PMID: 32654082 PMCID: PMC7353835 DOI: 10.1007/s10067-020-05275-1] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
The pathogenesis of Coronavirus disease 2019 (COVID-19) is gradually being comprehended. A high number of thrombotic episodes are reported, along with the mortality benefits of heparin. COVID-19 can be viewed as a prothrombotic disease. We overviewed the available evidence to explore this possibility. We identified various histopathology reports and clinical case series reporting thromboses in COVID-19. Also, multiple coagulation markers support this. COVID-19 can be regarded as a risk factor for thrombosis. Applying the principles of Virchow's triad, we described abnormalities in the vascular endothelium, altered blood flow, and platelet function abnormalities that lead to venous and arterial thromboses in COVID-19. Endothelial dysfunction, activation of the renin-angiotensin-aldosterone system (RAAS) with the release of procoagulant plasminogen activator inhibitor (PAI-1), and hyperimmune response with activated platelets seem to be significant contributors to thrombogenesis in COVID-19. Stratifying risk of COVID-19 thromboses should be based on age, presence of comorbidities, D-dimer, CT scoring, and various blood cell ratios. Isolated heparin therapy may not be sufficient to combat thrombosis in this disease. There is an urgent need to explore newer avenues like activated protein C, PAI-1 antagonists, and tissue plasminogen activators (tPA). These should be augmented with therapies targeting RAAS, antiplatelet drugs, repurposed antiinflammatory, and antirheumatic drugs. Key Points • Venous and arterial thromboses in COVID-19 can be viewed through the prism of Virchow's triad. • Endothelial dysfunction, platelet activation, hyperviscosity, and blood flow abnormalities due to hypoxia, immune reactions, and hypercoagulability lead to thrombogenesis in COVID-19. • There is an urgent need to stratify COVID-19 patients at risk for thrombosis using age, comorbidities, D-dimer, and CT scoring. • Patients with COVID-19 at high risk for thrombosis should be put on high dose heparin therapy.
Collapse
Affiliation(s)
- Sakir Ahmed
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar, India
| | - Olena Zimba
- Department of Internal Medicine No. 2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK) Russells Hall Hospital, Pensnett Road, Dudley, West Midlands DY1 2HQ UK
| |
Collapse
|
43
|
Huang TC, Wu HL, Chen SH, Wang YT, Wu CC. Thrombomodulin facilitates peripheral nerve regeneration through regulating M1/M2 switching. J Neuroinflammation 2020; 17:240. [PMID: 32799887 PMCID: PMC7477856 DOI: 10.1186/s12974-020-01897-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background Excessive inflammation within damaged tissue usually leads to delayed or insufficient regeneration, and nerves in the peripheral nervous system (PNS) generally do not recover fully following damage. Consequently, there is growing interest in whether modulation of the inflammatory response could help to promote nerve regeneration in the PNS. However, to date, there are no practical therapeutic strategies for manipulating inflammation after nerve injury. Thrombomodulin (TM) is a transmembrane glycoprotein containing five domains. The lectin-like domain of TM has the ability to suppress the inflammatory response. However, whether TM can modulate inflammation in the PNS during nerve regeneration has yet to be elucidated. Methods We investigated the role of TM in switching proinflammatory type 1 macrophages (M1) to anti-inflammatory type 2 macrophages (M2) in a human monocytic cell line (THP-1) and evaluated the therapeutic application of TM in transected sciatic nerve injury in rats. Results The administration of TM during M1 induction significantly reduced the expression levels of inflammatory cytokines, including TNF-a (p < 0.05), IL-6 (p < 0.05), and CD86 (p < 0.05), in THP-1 cells. Simultaneously, the expression levels of M2 markers, including IL-10 (p < 0.05) and CD206 (p < 0.05), were significantly increased in TM-treated THP-1 cells. Inhibition of IL-4R-c-Myc-pSTAT6-PPARγ signaling abolished the expression levels of IL-10 (p < 0.05) and CD206 (p < 0.05). The conditioned medium (CM) collected from M1 cells triggered an inflammatory response in primary Schwann cells, while CM collected from M1 cells treated with TM resulted in a dose-dependent reduction in inflammation. TM treatment led to better nerve regeneration when tested 6 weeks after injury and preserved effector muscle function. In addition, TM treatment reduced macrophage infiltration at the site of injury and led to potent M1 to M2 transition, thus indicating the anti-inflammatory capacity of TM. Conclusions Collectively, our findings demonstrate the anti-inflammatory role of TM during nerve regeneration. Therefore, TM represents a potential drug for the promotion and modulation of functional recovery in peripheral nerves that acts by regulating the M1/M2 ratio.
Collapse
Affiliation(s)
- Tzu-Chieh Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Rd, Tainan, 701, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Szu-Han Chen
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yun-Ting Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Rd, Tainan, 701, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan. .,Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
44
|
Hurtado DE, Erranz B, Lillo F, Sarabia-Vallejos M, Iturrieta P, Morales F, Blaha K, Medina T, Diaz F, Cruces P. Progression of regional lung strain and heterogeneity in lung injury: assessing the evolution under spontaneous breathing and mechanical ventilation. Ann Intensive Care 2020; 10:107. [PMID: 32761387 PMCID: PMC7407426 DOI: 10.1186/s13613-020-00725-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Protective mechanical ventilation (MV) aims at limiting global lung deformation and has been associated with better clinical outcomes in acute respiratory distress syndrome (ARDS) patients. In ARDS lungs without MV support, the mechanisms and evolution of lung tissue deformation remain understudied. In this work, we quantify the progression and heterogeneity of regional strain in injured lungs under spontaneous breathing and under MV. METHODS Lung injury was induced by lung lavage in murine subjects, followed by 3 h of spontaneous breathing (SB-group) or 3 h of low Vt mechanical ventilation (MV-group). Micro-CT images were acquired in all subjects at the beginning and at the end of the ventilation stage following induction of lung injury. Regional strain, strain progression and strain heterogeneity were computed from image-based biomechanical analysis. Three-dimensional regional strain maps were constructed, from which a region-of-interest (ROI) analysis was performed for the regional strain, the strain progression, and the strain heterogeneity. RESULTS After 3 h of ventilation, regional strain levels were significantly higher in 43.7% of the ROIs in the SB-group. Significant increase in regional strain was found in 1.2% of the ROIs in the MV-group. Progression of regional strain was found in 100% of the ROIs in the SB-group, whereas the MV-group displayed strain progression in 1.2% of the ROIs. Progression in regional strain heterogeneity was found in 23.4% of the ROIs in the SB-group, while the MV-group resulted in 4.7% of the ROIs showing significant changes. Deformation progression is concurrent with an increase of non-aerated compartment in SB-group (from 13.3% ± 1.6% to 37.5% ± 3.1%), being higher in ventral regions of the lung. CONCLUSIONS Spontaneous breathing in lung injury promotes regional strain and strain heterogeneity progression. In contrast, low Vt MV prevents regional strain and heterogeneity progression in injured lungs.
Collapse
Affiliation(s)
- Daniel E Hurtado
- Department of Structural and Geotechnical Engineering, School of Engineering Pontificia, Universidad Católica de Chile, Santiago, Chile.,Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile
| | - Benjamín Erranz
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Felipe Lillo
- Centro de Investigación de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Mauricio Sarabia-Vallejos
- Department of Structural and Geotechnical Engineering, School of Engineering Pontificia, Universidad Católica de Chile, Santiago, Chile
| | - Pablo Iturrieta
- Department of Structural and Geotechnical Engineering, School of Engineering Pontificia, Universidad Católica de Chile, Santiago, Chile
| | - Felipe Morales
- Centro de Investigación de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Katherine Blaha
- Pediatric Critical Care Unit, Clínica Alemana de Santiago, Santiago, Chile
| | - Tania Medina
- Pediatric Intensive Care Unit, Hospital El Carmen de Maipú, Santiago, Chile
| | - Franco Diaz
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile.,Pediatric Critical Care Unit, Clínica Alemana de Santiago, Santiago, Chile
| | - Pablo Cruces
- Centro de Investigación de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile. .,Pediatric Intensive Care Unit, Hospital El Carmen de Maipú, Santiago, Chile.
| |
Collapse
|
45
|
Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, He Y, Chopra A, Chieng HC, Tiwari A, Judson MA, Paulson B, Brademan DR, Zhu Y, Serrano LR, Linke V, Drake LA, Adam AP, Schwartz BS, Singer HA, Swanson S, Mosher DF, Stewart R, Coon JJ, Jaitovich A. Large-scale Multi-omic Analysis of COVID-19 Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.07.17.20156513. [PMID: 32743614 PMCID: PMC7388490 DOI: 10.1101/2020.07.17.20156513] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We performed RNA-Seq and high-resolution mass spectrometry on 128 blood samples from COVID-19 positive and negative patients with diverse disease severities. Over 17,000 transcripts, proteins, metabolites, and lipids were quantified and associated with clinical outcomes in a curated relational database, uniquely enabling systems analysis and cross-ome correlations to molecules and patient prognoses. We mapped 219 molecular features with high significance to COVID-19 status and severity, many involved in complement activation, dysregulated lipid transport, and neutrophil activation. We identified sets of covarying molecules, e.g., protein gelsolin and metabolite citrate or plasmalogens and apolipoproteins, offering pathophysiological insights and therapeutic suggestions. The observed dysregulation of platelet function, blood coagulation, acute phase response, and endotheliopathy further illuminated the unique COVID-19 phenotype. We present a web-based tool (covid-omics.app) enabling interactive exploration of our compendium and illustrate its utility through a comparative analysis with published data and a machine learning approach for prediction of COVID-19 severity.
Collapse
Affiliation(s)
- Katherine A. Overmyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ian J. Miller
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Joseph Balnis
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | | | - Trenton M. Peters-Clarke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Jesse G. Meyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Qiuwen Quan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Laura K. Muehlbauer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Edna A. Trujillo
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yuchen He
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Amit Chopra
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Hau C. Chieng
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Anupama Tiwari
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Division of Sleep Medicine, Albany Medical Center, Albany, NY, USA
| | - Marc A. Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Brett Paulson
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Dain R. Brademan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yunyun Zhu
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lia R. Serrano
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Vanessa Linke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lisa A. Drake
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Alejandro P. Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Department of Ophthalmology, Albany Medical College, Albany, NY, USA
| | | | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Scott Swanson
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Deane F. Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Joshua J. Coon
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Morgridge Institute for Research, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
46
|
Kreyer S, Baker WL, Scaravilli V, Linden K, Belenkiy SM, Necsoiu C, Muders T, Putensen C, Chung KK, Cancio LC, Batchinsky AI. Assessment of spontaneous breathing during pressure controlled ventilation with superimposed spontaneous breathing using respiratory flow signal analysis. J Clin Monit Comput 2020; 35:859-868. [PMID: 32535849 PMCID: PMC7293172 DOI: 10.1007/s10877-020-00545-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/06/2020] [Indexed: 11/25/2022]
Abstract
Integrating spontaneous breathing into mechanical ventilation (MV) can speed up liberation from it and reduce its invasiveness. On the other hand, inadequate and asynchronous spontaneous breathing has the potential to aggravate lung injury. During use of airway-pressure-release-ventilation (APRV), the assisted breaths are difficult to measure. We developed an algorithm to differentiate the breaths in a setting of lung injury in spontaneously breathing ewes. We hypothesized that differentiation of breaths into spontaneous, mechanical and assisted is feasible using a specially developed for this purpose algorithm. Ventilation parameters were recorded by software that integrated ventilator output variables. The flow signal, measured by the EVITA® XL (Lübeck, Germany), was measured every 2 ms by a custom Java-based computerized algorithm (Breath-Sep). By integrating the flow signal, tidal volume (VT) of each breath was calculated. By using the flow curve the algorithm separated the different breaths and numbered them for each time point. Breaths were separated into mechanical, assisted and spontaneous. Bland Altman analysis was used to compare parameters. Comparing the values calculated by Breath-Sep with the data from the EVITA® using Bland-Altman analyses showed a mean bias of - 2.85% and 95% limits of agreement from - 25.76 to 20.06% for MVtotal. For respiratory rate (RR) RRset a bias of 0.84% with a SD of 1.21% and 95% limits of agreement from - 1.53 to 3.21% were found. In the cluster analysis of the 25th highest breaths of each group RRtotal was higher using the EVITA®. In the mechanical subgroup the values for RRspont and MVspont the EVITA® showed higher values compared to Breath-Sep. We developed a computerized method for respiratory flow-curve based differentiation of breathing cycle components during mechanical ventilation with superimposed spontaneous breathing. Further studies in humans and optimizing of this technique is necessary to allow for real-time use at the bedside.
Collapse
Affiliation(s)
- Stefan Kreyer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany.
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA.
| | - William L Baker
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
| | - Vittorio Scaravilli
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milano, MI, Italy
| | - Katharina Linden
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
- Pediatric Department, University Hospital Bonn, Bonn, Germany
| | - Slava M Belenkiy
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
- Department of Anesthesiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Corina Necsoiu
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
| | - Thomas Muders
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Kevin K Chung
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Leopoldo C Cancio
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
| | - Andriy I Batchinsky
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
- The Geneva Foundation, Tacoma, WA, USA
| |
Collapse
|
47
|
Jacobs MS, Loef B, Reidinga AC, Postma MJ, Van Hulst M, Tieleman RG. Incidence, treatment and mortality of new-onset atrial fibrillation patients at the intensive care unit. Open Heart 2020; 7:e001226. [PMID: 32509315 PMCID: PMC7254104 DOI: 10.1136/openhrt-2019-001226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/27/2020] [Accepted: 02/20/2020] [Indexed: 01/01/2023] Open
Abstract
Objective Critically ill patients admitted to the intensive care unit (ICU) often develop atrial fibrillation (AF), with an incidence of around 5%. Stroke prevention in AF is well described in clinical guidelines. The extent to which stroke prevention is prescribed to ICU patients with AF is unknown. We aimed to determine the incidence of new-onset AF and describe stroke prevention strategies initiated on the ICU of our teaching hospital. Also, we compared mortality in patients with new-onset AF to critically ill patients with previously diagnosed AF and patients without any AF. Methods This study was a retrospective cohort study including all admissions to the ICU of the Martini Hospital (Groningen, The Netherlands) in the period 2011 to 2016. Survival analyses were performed using these real-world data. Results In total, 3334 patients were admitted to the ICU, of whom 213 patients (6.4%) developed new-onset AF. 583 patients (17.5%) had a previous AF diagnosis, the other patients were in sinus rhythm. In-hospital mortality and 1-year mortality after hospital discharge were significantly higher for new-onset AF patients compared with patients with no history of AF or previously diagnosed AF. At hospital discharge, only 56.3% of the new-onset AF-patients eligible for stroke prevention received an anticoagulant. Anticoagulation was not dependent on CHA2DS2-VASc score or other patient characteristics. An effect of anticoagulative status on mortality was not significant. Conclusion AF is associated with increased mortality in critically ill patients admitted to the ICU. More guidance is needed to optimise anticoagulant treatment in critically ill new-onset AF patients.
Collapse
Affiliation(s)
- Maartje S Jacobs
- Department of Clinical Pharmacy and Toxicology, Martini Hospital, Groningen, The Netherlands.,Department of Pharmacy, University of Groningen, Groningen Research Institute of Pharmacy, Groningen, Netherlands.,Department of Health Sciences, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Bert Loef
- Department of Intensive Care, Martini Hospital, Groningen, Netherlands
| | - Auke C Reidinga
- Department of Intensive Care, Martini Hospital, Groningen, Netherlands
| | - Maarten J Postma
- Department of Pharmacy, University of Groningen, Groningen Research Institute of Pharmacy, Groningen, Netherlands.,Department of Health Sciences, University of Groningen, University Medical Center, Groningen, The Netherlands.,Department of Economics, Econometrics & Business, University of Groningen, Faculty of Economics & Business, Groningen, Netherlands
| | - Marinus Van Hulst
- Department of Clinical Pharmacy and Toxicology, Martini Hospital, Groningen, The Netherlands.,Department of Health Sciences, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Robert G Tieleman
- Department of Cardiology, Martini Hospital, Groningen, Netherlands.,Department of Cardiology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
48
|
Tombling BJ, Wang CK, Craik DJ. EGF‐artige und andere disulfidreiche Mikrodomänen als therapeutische Molekülgerüste. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201913809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Benjamin J. Tombling
- Institute for Molecular Bioscience The University of Queensland Brisbane QLD 4072 Australien
| | - Conan K. Wang
- Institute for Molecular Bioscience The University of Queensland Brisbane QLD 4072 Australien
| | - David J. Craik
- Institute for Molecular Bioscience The University of Queensland Brisbane QLD 4072 Australien
| |
Collapse
|
49
|
Tombling BJ, Wang CK, Craik DJ. EGF-like and Other Disulfide-rich Microdomains as Therapeutic Scaffolds. Angew Chem Int Ed Engl 2020; 59:11218-11232. [PMID: 31867866 DOI: 10.1002/anie.201913809] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Indexed: 12/20/2022]
Abstract
Disulfide bonds typically introduce conformational constraints into peptides and proteins, conferring improved biopharmaceutical properties and greater therapeutic potential. In our opinion, disulfide-rich microdomains from proteins are potentially a rich and under-explored source of drug leads. A survey of the UniProt protein database shows that these domains are widely distributed throughout the plant and animal kingdoms, with the EGF-like domain being the most abundant of these domains. EGF-like domains exhibit large diversity in their disulfide bond topologies and calcium binding modes, which we classify in detail here. We found that many EGF-like domains are associated with disease phenotypes, and the interactions they mediate are potential therapeutic targets. Indeed, EGF-based therapeutic leads have been identified, and we further propose that these domains can be optimized to expand their therapeutic potential using chemical design strategies. This Review highlights the potential of disulfide-rich microdomains as future peptide therapeutics.
Collapse
Affiliation(s)
- Benjamin J Tombling
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Conan K Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
50
|
Kawano N, Wada H, Uchiyama T, Kawasugi K, Madoiwa S, Takezako N, Suzuki K, Seki Y, Ikezoe T, Hattori T, Okamoto K. Analysis of the association between resolution of disseminated intravascular coagulation (DIC) and treatment outcomes in post-marketing surveillance of thrombomodulin alpha for DIC with infectious disease and with hematological malignancy by organ failure. Thromb J 2020; 18:2. [PMID: 32047363 PMCID: PMC7006199 DOI: 10.1186/s12959-020-0216-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/28/2020] [Indexed: 12/29/2022] Open
Abstract
Background Although disseminated intravascular coagulation (DIC) is life-threatening, any organ failure associated with DIC resolution and outcomes have been unclear. Patients and methods A total of 2795 DIC patients (infection: 1990, hematological malignancy: 805) were analyzed in the post-marketing surveillance of thrombomodulin alpha (TM-α). The background factors of sequential organ failure assessment (SOFA) and antithrombin (AT) were investigated in DIC with infectious disease for their association with DIC resolution and outcome using κ statistics, indicating DIC resolution and survival or DIC non-resolution and non-survival. The same analyses were performed for total bilirubin, creatinine, lactate dehydrogenase, and underlying disease in DIC with hematological malignancy. Results In DIC with infectious disease, higher SOFA score severity was closely correlated with lower overall survival in both the DIC resolution and non-resolution groups, but AT activity was not. κ coefficients were 0.234, 0.295, and 0.311 for the SOFA score 0-6, 7-12, and 13-24 groups, respectively. In DIC with hematological malignancy, κ coefficients of total bilirubin were 0.251 and 0.434, and those of creatinine were 0.283 and 0.437 in the normal and abnormal groups, respectively, showing better concordance in the abnormal group than in the normal. Other factors had poor concordance. Conclusion In DIC with infectious disease, DIC resolution is an important therapeutic target in patients who have higher SOFA score severity. In DIC with hematological malignancy, DIC resolution is similarly important in patients with abnormality of bilirubin and/or creatinine. Trial registration The clinical characteristics and treatment outcomes of patients with DIC treated with TM-α between May 2008 and April 2010 were retrospectively analyzed by subgroup analysis of the post-marketing surveillance data.
Collapse
Affiliation(s)
- Noriaki Kawano
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, 5-30 Kitatakamatsu, Miyazaki, 880-8510 Japan
| | - Hideo Wada
- Department of General Medicine, Mie Prefectural General Medical Center, Mie, Japan
| | - Toshimasa Uchiyama
- Department of Laboratory Medicine, National Hospital Organization Takasaki General Medical Center, Gunma, Japan
| | - Kazuo Kawasugi
- 4Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Seiji Madoiwa
- 5Department of Clinical Laboratory Medicine, Tokyo Saiseikai Central Hospital, Tokyo, Japan
| | - Naoki Takezako
- 6Department of Hematology, National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Kei Suzuki
- 7Emergency and Critical Care Center, Mie University Hospital, Mie, Japan
| | - Yoshinobu Seki
- 8Department of Hematology, Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Takayuki Ikezoe
- 9Department of Hematology, Fukushima Medical University, Fukushima, Japan
| | - Tsuyoshi Hattori
- 10Department of Medical Affairs, Asahi Kasei Pharma Corporation, Tokyo, Japan
| | - Kohji Okamoto
- Department of Surgery, Center for Gastroenterology and Liver Disease, Kitakyushu City Yahata Hospital, Fukuoka, Japan
| |
Collapse
|