1
|
Coloretti I, Corcione A, De Pascale G, Donati A, Forfori F, Marietta M, Panigada M, Simioni P, Tascini C, Viale P, Girardis M. Protein C in adult patients with sepsis: from pathophysiology to monitoring and supplementation. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2025; 5:21. [PMID: 40229903 PMCID: PMC11998338 DOI: 10.1186/s44158-025-00243-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Protein C (PC) plays a crucial role in modulating inflammation and coagulation in sepsis. Its anticoagulant and cytoprotective properties are critical in mitigating sepsis-induced coagulopathy, which is associated with high mortality rates. In sepsis, low levels of PC are associated with an elevated risk of multiple organ dysfunction and increased mortality. Routine monitoring of PC levels is not widely implemented but appears relevant in selected populations, such as patients with purpura fulminans, sepsis-induced coagulopathy (SIC), disseminated intravascular coagulopathy (DIC) or hyperinflammatory septic shock phenotypes. Treatment with PC has been limited to PC concentrate approved for paediatric use in congenital PC deficiencies and purpura fulminans, while the efficacy of PC supplementation in sepsis remains a subject of debate. Considering the physiological significance of PC and its role in sepsis pathophysiology, additional studies are necessary to fully elucidate its therapeutic efficacy in specific clinical settings.
Collapse
Affiliation(s)
- Irene Coloretti
- Anaesthesiology and Intensive Care Department, University Hospital of Modena, University of Modena, Reggio Emilia, Modena, Italy.
| | - Antonio Corcione
- Department of Critical Care, AORN Ospedali Dei Colli, Naples, Italy
| | - Gennaro De Pascale
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Fondazione Policlinico Universitario A. Gemelli IRCCS, Anestesiologiche E Della Rianimazione, Rome, Italy
| | - Abele Donati
- Department of Biomedical Sciences and Public Health, Università Politecnica Delle Marche, Ancona, Italy
- Anesthesia and Intensive Care, Azienda Ospedaliero Universitaria Delle Marche, Ancona, Italy
| | - Francesco Forfori
- Dipartimento Di Patologia Chirurgica, Medica, Molecolare Ed Area Critica, Università Di Pisa. AOUP, Pisa, Italy
| | - Marco Marietta
- Department of Hematology-Azienda Ospedaliero, Universitaria Di Modena, Modena, Italy
| | - Mauro Panigada
- Department of Anesthesia, Intensive Care and Emergency, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Simioni
- Clinica Medica 1, Azienda Ospedale Università Di Padova, Padua, Italy
| | - Carlo Tascini
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Infectious Diseases Clinic, ASUFC "Santa Maria Della Misericordia" University Hospital of Udine, Udine, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Massimo Girardis
- Anaesthesiology and Intensive Care Department, University Hospital of Modena, University of Modena, Reggio Emilia, Modena, Italy
| |
Collapse
|
2
|
Bendapudi PK, Losman JA. How I diagnose and treat acute infection-associated purpura fulminans. Blood 2025; 145:1358-1368. [PMID: 39786416 DOI: 10.1182/blood.2024025078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Purpura fulminans (PF) is a rare but devastating complication of sepsis characterized by a highly thrombotic subtype of disseminated intravascular coagulation (DIC). A medical emergency, PF often requires the involvement of consultant hematologists to assist with diagnosis and management of patients who are in a highly dynamic and deteriorating clinical situation. Patients who survive past the first 24 to 72 hours often die from complications of unchecked thrombosis rather than shock, and survivors are usually left with severe scarring and tissue loss. Despite these challenging features, PF is a pathophysiologically distinct, homogeneous, and highly predictable form of sepsis-associated DIC for which poor outcomes are not a foregone conclusion. The fundamental pathologic lesion in PF is a failure of the anticoagulant protein C pathway, which leads to uncontrolled microvascular clotting and inadequate protein C-mediated cytoprotective effects, which are vital for survival in sepsis. Herein, we review the clinical features and diagnosis of PF. Drawing from existing clinical literature and recent advances in our understanding of the pathophysiology of PF, we describe rationally designed treatment approaches for this disorder, including repletion of natural circulating anticoagulants, use of therapeutic anticoagulation, and ways to optimize transfusion support, and we outline specific interventions that we would recommend avoiding.
Collapse
Affiliation(s)
- Pavan K Bendapudi
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Center for the Development of Therapeutics, The Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Julie-Aurore Losman
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
3
|
Silva BRDS, Sidarta-Oliveira D, Morari J, Bombassaro B, Jara CP, Simeoni CL, Parise PL, Proenca-Modena JL, Velloso LA, Velander WH, Araújo EP. Protein C Pretreatment Protects Endothelial Cells from SARS-CoV-2-Induced Activation. Viruses 2024; 16:1049. [PMID: 39066212 PMCID: PMC11281670 DOI: 10.3390/v16071049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
SARS-CoV-2 can induce vascular dysfunction and thrombotic events in patients with severe COVID-19; however, the cellular and molecular mechanisms behind these effects remain largely unknown. In this study, we used a combination of experimental and in silico approaches to investigate the role of PC in vascular and thrombotic events in COVID-19. Single-cell RNA-sequencing data from patients with COVID-19 and healthy subjects were obtained from the publicly available Gene Expression Omnibus (GEO) repository. In addition, HUVECs were treated with inactive protein C before exposure to SARS-CoV-2 infection or a severe COVID-19 serum. An RT-qPCR array containing 84 related genes was used, and the candidate genes obtained were evaluated. Activated protein C levels were measured using an ELISA kit. We identified at the single-cell level the expression of several pro-inflammatory and pro-coagulation genes in endothelial cells from the patients with COVID-19. Furthermore, we demonstrated that exposure to SARS-CoV-2 promoted transcriptional changes in HUVECs that were partly reversed by the activated protein C pretreatment. We also observed that the serum of severe COVID-19 had a significant amount of activated protein C that could protect endothelial cells from serum-induced activation. In conclusion, activated protein C protects endothelial cells from pro-inflammatory and pro-coagulant effects during exposure to the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Bruna Rafaela dos Santos Silva
- School of Nursing, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - Joseane Morari
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - Bruna Bombassaro
- School of Nursing, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | | | - Camila Lopes Simeoni
- Laboratory of Emerging Viruses, Institute of Biology, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; (C.L.S.); (P.L.P.); (J.L.P.-M.)
| | - Pierina Lorencini Parise
- Laboratory of Emerging Viruses, Institute of Biology, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; (C.L.S.); (P.L.P.); (J.L.P.-M.)
| | - José Luiz Proenca-Modena
- Laboratory of Emerging Viruses, Institute of Biology, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; (C.L.S.); (P.L.P.); (J.L.P.-M.)
| | - Licio A. Velloso
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - William H. Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE 68588, USA;
| | - Eliana P. Araújo
- School of Nursing, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| |
Collapse
|
4
|
Levy-Mendelovich S, Cohen O, Klang E, Kenet G. 50 Years of Pediatric Hemostasis: Knowledge, Diagnosis, and Treatment. Semin Thromb Hemost 2023; 49:217-224. [PMID: 36174607 DOI: 10.1055/s-0042-1756704] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Studies from the past 50 years have contributed to the expanding knowledge regarding developmental hemostasis. This is a dynamic process that begins in the fetal phase and is characterized by physiological variations in platelet counts and function, and concentrations of most coagulation factors and the native coagulation inhibitors in early life, as compared with adulthood. The developmental hemostasis studies since the 1980 to 1990s established the laboratory reference values for coagulation factors. It was only a decade or two later, that thromboelastography (TEG) or (rotational thromboelastometry [ROTEM]) as well as thrombin generation studies, provided special pediatric reference values along with the ability to evaluate clot formation and lysis. In addition, global whole blood-based clotting assays provided point of care guidance for proper transfusion support to children hospitalized in intensive care units or undergoing surgery. Although uncommon, thrombosis in children and neonates is gaining increasing recognition, typically as a secondary complication in sick children. Bleeding in children, and particularly intracerebral hemorrhage in newborns, still represent a therapeutic challenge. Notably, our review will outline the advancements in understanding developmental hemostasis and its manifestations, with respect to the pathophysiology of thrombosis and bleeding complications in young children. The changes of transfusion policy and approach to thrombophilia testing during the last decade will be mentioned. Subsequently, a brief summary of the data on anticoagulant treatments in pediatric patients will be presented. Finally, we will point out the 10 most cited articles in the field of pediatric and neonatal hemostasis.
Collapse
Affiliation(s)
- Sarina Levy-Mendelovich
- National Hemophilia Center, Coagulation Unit and Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omri Cohen
- National Hemophilia Center, Coagulation Unit and Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eyal Klang
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Israel
| | - Gili Kenet
- National Hemophilia Center, Coagulation Unit and Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
5
|
Contou D, Urbina T, de Prost N. Understanding purpura fulminans in adult patients. Intensive Care Med 2022; 48:106-110. [PMID: 34846563 DOI: 10.1007/s00134-021-06580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/08/2021] [Indexed: 01/15/2023]
Affiliation(s)
- Damien Contou
- Service de Réanimation Polyvalente, Centre Hospitalier Victor Dupouy, 69, Rue du Lieutenant-Colonel Prud'hon, 95100, Argenteuil, France.
| | - Tomas Urbina
- Service de Médecine Intensive Réanimation, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nicolas de Prost
- Service de Médecine Intensive Réanimation, Groupe de Recherche CARMAS, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| |
Collapse
|
6
|
Massaud-Ribeiro L, Silami PHNC, Lima-Setta F, Prata-Barbosa A. Pediatric Sepsis Research: Where Are We and Where Are We Going? Front Pediatr 2022; 10:829119. [PMID: 35223703 PMCID: PMC8873512 DOI: 10.3389/fped.2022.829119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/20/2022] [Indexed: 01/15/2023] Open
Abstract
Sepsis continues to be one of the leading causes of admission to the Pediatric Intensive Care Unit, representing a great challenge for researchers and healthcare staff. This mini review aims to assess research on pediatric sepsis over the years. Of the 2,698 articles retrieved from the Scopus database, the 100 most cited were selected (50 published since 2000 and 50 published since 2016). The most cited studies, published in the 21st century, are highlighted, with their main findings and perspectives.
Collapse
Affiliation(s)
- Letícia Massaud-Ribeiro
- Pediatric Intensive Care Unit, Department of Pediatrics, Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Pediatric Intensive Care Unit, Department of Pediatrics, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Pedro Henrique Nunes Costa Silami
- Pediatric Intensive Care Unit, Department of Pediatrics, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Pediatric Intensive Care Unit, Department of Pediatrics, Hospital Estadual da Criança, Rio de Janeiro, Brazil
| | - Fernanda Lima-Setta
- Pediatric Intensive Care Unit, Department of Pediatrics, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Department of Pediatrics, Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Arnaldo Prata-Barbosa
- Department of Pediatrics, Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Detectable A Disintegrin and Metalloproteinase With Thrombospondin Motifs-1 in Serum Is Associated With Adverse Outcome in Pediatric Sepsis. Crit Care Explor 2021; 3:e0569. [PMID: 34765980 PMCID: PMC8577672 DOI: 10.1097/cce.0000000000000569] [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: 11/25/2022] Open
Abstract
Supplemental Digital Content is available in the text. A Disintegrin and Metalloproteinase with Thrombospondin Motifs-1 is hypothesized to play a role in the pathogenesis of invasive infection, but studies in sepsis are lacking.
Collapse
|
8
|
Zhao R, Lang TC, Kim A, Wijewardena A, Vandervord J, McGrath R, Fulcher G, Xue M, Jackson C. Early protein C activation is reflective of burn injury severity and plays a critical role in inflammatory burden and patient outcomes. Burns 2021; 48:91-103. [PMID: 34175158 DOI: 10.1016/j.burns.2021.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Navigating the complexities of a severe burn injury is a challenging endeavour where the natural course of some patients can be difficult to predict. Straddling both the coagulation and inflammatory cascades that feature strongly in the burns systemic pathophysiology, we propose the pleiotropic protein C (PC) system may produce a viable biomarker to assist traditional evaluation methods for diagnostic and prognostic purposes. METHODS We enrolled 86 patients in a prospective observational cohort study. Over three weeks, serial blood samples were taken and measured for PC, activated (A)PC, their receptor endothelial protein C receptor (EPCR), and a panel of inflammatory cytokines including C-reactive protein (CRP), tumour necrosis factor-α, interleukin (IL)-1β, IL-6, IL-8, and IL-17. Their temporal trends were analysed alongside clinical factors including burn size, burn depth, presence of inhalational injury, and a composite outcome of requiring increased support. RESULTS (i) APC increased from a nadir on Day 3 (2.3±2.1ng/mL vs 4.1±2.5ng/mL by Day 18, p<0.0005), only becoming appropriately correlated to PC from Day 6 onwards (r=0.412-0.721, p<0.05 for all Days 6-21). (ii) This early disturbance in the PC system was amplified in the more severe burns (≥30% total body surface area, predominantly full thickness, or with inhalational injury), which were characterised by a marked fall in PC activation (approximated by APC/PC ratio) and APC levels during Days 0-3 with low unchanged PC levels. Critically low levels of this cytoprotective agent was associated with greater inflammatory burden, as reflected by significantly elevated CRP, IL-6, and IL-8 levels in the more severe compared to less severe burns, and by negative correlations between both PC and APC with most inflammatory cytokines. (iii) Alongside clinical markers of severity at admission (burn size, burn depth, and presence of inhalational injury), only Day 0 APC/PC ratio (OR 1.048 (1.014-1.083), p=0.006), APC (OR 1.364 (1.032-1.803), p=0.029), PC (OR 0.899 (0.849-0.953), p<0.0005), and not any inflammatory cytokines were predictive markers of requiring increased support. Uniquely, decreased Day 0 PC was further individually associated with each increased total length of stay, ICU length of stay, intravenous fluid resuscitation, and total surgeries, as well as possibly mortality. CONCLUSION An early functional depletion of the cytoprotective PC system provides a physiological link between severe burns and the cytokine storm, likely contributing to worse outcomes. Our findings on the changes in APC, PC and PC activation during this pathological state support APC and PC as early diagnostic and prognostic biomarkers, and provides a basis for their therapeutic potential in severe burn injuries.
Collapse
Affiliation(s)
- Ruilong Zhao
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| | - Thomas Charles Lang
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Albert Kim
- Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | | | - John Vandervord
- Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Rachel McGrath
- Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Gregory Fulcher
- Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Meilang Xue
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Christopher Jackson
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| |
Collapse
|
9
|
Bendapudi PK, Whalen MJ, Lahoud-Rahme M, Villalba JA. Case 7-2021: A 19-Year-Old Man with Shock, Multiple Organ Failure, and Rash. N Engl J Med 2021; 384:953-963. [PMID: 33704941 DOI: 10.1056/nejmcpc2027093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Pavan K Bendapudi
- From the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Massachusetts General Hospital, and the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Harvard Medical School - both in Boston
| | - Michael J Whalen
- From the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Massachusetts General Hospital, and the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Harvard Medical School - both in Boston
| | - Manuella Lahoud-Rahme
- From the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Massachusetts General Hospital, and the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Harvard Medical School - both in Boston
| | - Julian A Villalba
- From the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Massachusetts General Hospital, and the Departments of Medicine (P.K.B.), Pediatrics (M.J.W., M.L.-R.), and Pathology (J.A.V.), Harvard Medical School - both in Boston
| |
Collapse
|
10
|
Abstract
Thrombocytopenia-associated multiple organ failure is a clinical phenotype encompassing a spectrum of syndromes associated with disseminated microvascular thromboses. Autopsies performed in patients that died with thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, or disseminated intravascular coagulation reveal specific findings that can differentiate these 3 entities. Significant advancements have been made in our understanding of the pathologic mechanisms of these syndromes. Von Willebrand factor and ADAMTS-13 play a central role in thrombotic thrombocytopenic purpura. Shiga toxins and the complement pathway drive the hemolytic uremic syndrome pathology. Tissue factor activity is vital in the development of disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Trung C Nguyen
- Department of Pediatrics, Critical Care Medicine Section, Texas Children's Hospital/Baylor College of Medicine, 6651 Main Street, MC: E 1420, Houston, TX 77030, USA; The Center for Translational Research on Inflammatory Diseases (CTRID), The Michael E. DeBakey Veteran Administration Medical Center, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Djurdjevic N, Taweesedt PT, Paulson M, LaNou A, Radovanovic M, Patel JN, Veselinovic M, McDermott WR, Dumic I. Septic Shock and Purpura Fulminans Due to Streptococcus pneumoniae Bacteremia in an Unvaccinated Immunocompetent Adult: Case Report and Review. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e923266. [PMID: 32513908 PMCID: PMC7304654 DOI: 10.12659/ajcr.923266] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/12/2020] [Accepted: 04/03/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Despite proven efficacy of vaccinations against Streptococcus pneumoniae in preventing infection, only 70% of eligible individuals receive the vaccine in the United States. Pneumococcal bacteremia represents a form of invasive pneumococcal disease and is associated with high mortality, especially in immunocompromised patients and the elderly. Purpura fulminans is a rare complication and manifestation of disseminated intravascular coagulation and sepsis. It is exceedingly rare in the setting of pneumococcal bacteremia, particularly in immunocompetent individuals. CASE REPORT We report a generally healthy 67-year-old male with schizophrenia who refused pneumococcal vaccination. He had an intact and functional spleen with a functional immune system. The patient presented with fever and diarrhea. He subsequently progressed to develop purpura fulminans and septic shock due to S. pneumoniae bacteremia. Despite an extensive search for the primary source of infection, none could not be identified. Due to timely initiation of appropriate antibiotic therapy and aggressive supportive care in an intensive care unit, he recovered despite multi-organ failure that developed throughout his hospitalization. CONCLUSIONS We present a rare manifestation of a potentially preventable disease and emphasize the importance of pneumococcal vaccination in order to decrease the risk of developing invasive pneumococcal disease. Furthermore, we discuss etiology, diagnosis, differential diagnosis, and evidence-based management of purpura fulminans and invasive pneumococcal disease with a literature review. Purpura fulminans due to S. pneumoniae is exceedingly rare in immunocompetent patients and an unusual clinical manifestation of pneumococcal bacteremia.
Collapse
Affiliation(s)
| | | | | | - Abigail LaNou
- Mayo Clinic, Alix School of Medicine, Rochester, MN, U.S.A
| | | | - Janki N. Patel
- Mayo Clinic, Alix School of Medicine, Rochester, MN, U.S.A
| | | | | | - Igor Dumic
- Mayo Clinic, Alix School of Medicine, Rochester, MN, U.S.A
| |
Collapse
|
12
|
Persistence of endothelial thrombomodulin in a patient with infectious purpura fulminans treated with protein C concentrate. Blood Adv 2019; 2:2917-2921. [PMID: 30396911 DOI: 10.1182/bloodadvances.2018024430] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/11/2018] [Indexed: 12/26/2022] Open
|
13
|
Kumar A, Shah NP, Menon V, Nissen SE. Purpura fulminans manifesting with Staphylococcus aureus endocarditis: a case report. Eur Heart J Case Rep 2019; 3:5498064. [PMID: 31449632 PMCID: PMC6601163 DOI: 10.1093/ehjcr/ytz077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/28/2018] [Accepted: 04/26/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Purpura fulminans (PF) is a haematologic emergency that can occur in the setting of severe septic shock. Its pathophysiology is not well-understood; however, some evidence suggests it may be mediated by excessive protein C consumption. CASE SUMMARY In this case report, we describe a patient with PF secondary to methicillin-resistant Staphylococcus aureus endocarditis. She presented with severe septic shock and, despite haemodynamic improvement, developed a significant purpuric rash. Diagnostic work-up was notable for severely decreased serum levels of protein C. This patient was successfully treated with protein C concentrate and surgical valve replacement. DISCUSSION While PF is rarely associated with S. aureus infection, this presentation may be more frequently encountered among clinicians in the current opioid epidemic. Quick recognition is crucial and a multidisciplinary approach, including intravenous infusion of protein C, may be considered.
Collapse
Affiliation(s)
- Anirudh Kumar
- Heart and Vascular Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Desk J3-5, Cleveland, OH, USA
- Corresponding author. Tel: +1 216 970 7715,
| | - Nishant P Shah
- Heart and Vascular Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Desk J3-5, Cleveland, OH, USA
| | - Venu Menon
- Heart and Vascular Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Desk J3-5, Cleveland, OH, USA
| | - Steven E Nissen
- Heart and Vascular Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Desk J3-5, Cleveland, OH, USA
| |
Collapse
|
14
|
Hartman SJF, Boeddha NP, Ekinci E, Koch BCP, Donders R, Hazelzet JA, Driessen GJ, de Wildt SN. Target attainment of cefotaxime in critically ill children with meningococcal septic shock as a model for cefotaxime dosing in severe pediatric sepsis. Eur J Clin Microbiol Infect Dis 2019; 38:1255-1260. [PMID: 30968258 PMCID: PMC6570664 DOI: 10.1007/s10096-019-03535-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 12/21/2022]
Abstract
Reduced target attainment of β-lactam antibiotics is reported in critically ill patients. However, as target attainment of cefotaxime in severely ill pediatric sepsis patients may differ from adults due to age-related variation in pharmacokinetics, we aimed to assess target attainment of cefotaxime in this pilot study using meningococcal septic shock patients as a model for severe sepsis. Secondary analysis of prospectively collected data from a randomized controlled trial. Children with meningococcal septic shock (1 month to 18 years) included in this study received cefotaxime 100-150 mg/kg/day as antibiotic treatment. Left-over plasma samples were analyzed using LC-MS/MS to determine cefotaxime concentrations. MIC values from EUCAST were used to determine target attainment of cefotaxime for Neisseria meningitidis (0.125 mg/l), but also for Streptococcus pneumoniae (0.5 mg/l), Enterobacteriaceae (1 mg/l), and Staphylococcus aureus (4 mg/l). Target attainment was adequate when all samples exceeded MIC or fourfold MIC values. One thirty-six plasma samples of 37 severe septic shock patients were analyzed for cefotaxime concentrations. Median age was 2 years with a median PRISM-score of 24 and mortality of 24.8%. The median unbound cefotaxime concentration was 4.8 mg/l (range 0-48.7). Target attainment ranged from 94.6% for the MIC of N. meningitidis to 16.2% for fourfold the MIC S. aureus. Creatinine levels were significantly correlated with cefotaxime levels. Target attainment of cefotaxime with current dosing guidelines seems to be adequate for N. meningitidis but seems to fail for more frequently encountered pathogens in severely ill children.
Collapse
Affiliation(s)
- Stan J F Hartman
- Department of Pharmacology-Toxicology, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| | - Navin P Boeddha
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ebru Ekinci
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Pharmacy, ErasmusMC, Rotterdam, The Netherlands
| | - Rogier Donders
- Department for Health Evidence, Radboudumc, Nijmegen, The Netherlands
| | - Jan A Hazelzet
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gertjan J Driessen
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Juliana Children's Hospital, Haga Teaching Hospital, The Hague, The Netherlands
| | - Saskia N de Wildt
- Department of Pharmacology-Toxicology, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.,Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Gao TY, Yang WC, Zhou FH, Song Q. Cooling therapy upregulates protein C activation in heat stressed rats: An experimental study. J Cell Physiol 2019; 234:14181-14186. [PMID: 30633351 DOI: 10.1002/jcp.28114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/21/2018] [Indexed: 11/09/2022]
Abstract
Protein C (PC) pathway homeostasis is implicated in heat stress (HS). This study determines whether cooling could improve the PC pathway in HS. Fifty-six anesthetized rats were warmed to achieve HS (rectal temperature [Tr] 42°C). These rats were divided into seven groups: (a) control group:sacrifice immediately 15 min after HS; (b) HS+I:sacrifice immediately after 15 min ice-water treatment or (c) 3 hr after HS; (d) HS+C:sacrifice immediately after 15-min cold-water treatment or (e) 3 hr after HS; (f) HS: sacrifice immediately 15 min after HS or (g) 3 hr after HS. Plasma PC, activated protein C (APC), and soluble thrombomodulin (sTM) levels were tested at both time points. After cooling, Tr in the HS+I and HS+C groups significantly decreased, when compared with the HS group, and Tr was significantly lower in the HS+I group than in the HS+C group ( p < 0.05). Furthermore, sTM levels were highest in the HS group among the groups at both time points. Plasma PC and APC levels increased after HS. In the HS+I and HS+C groups, plasma APC levels and the APC/PC ratio significantly increased at both time points. The proportions were significantly higher in the HS+I group than in the HS+C group, and there was no significant increase in APC/PC ratio in the HS group. Cooling exerts an anticoagulant effect following HS by increasing APC levels. Ice-water blanket therapy is more effective than cold-water blanket therapy in increasing APC levels.
Collapse
Affiliation(s)
- Tie-Ying Gao
- Department of Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Wen-Chao Yang
- Department of Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fei-Hu Zhou
- Department of Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qing Song
- Department of Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
16
|
Abstract
Neonatal purpura fulminans (PF) is a life-threatening disorder caused by congenital or acquired deficiencies of protein C (PC) or S. PF presents as a cutaneous manifestation of disseminated intravascular coagulation. We describe a case of PF in a newborn with left leg ischemia and undetectable PC levels soon after birth. Despite anticoagulation therapy and PC concentrate, left foot amputation was required. Genetic testing of PROC for congenital PC deficiency was normal. This case highlights the course of PF due to acquired PC deficiency in a newborn treated with PC concentrate which is rarely described in the literature.
Collapse
|
17
|
The evolution of activated protein C plasma levels in septic shock and its association with mortality: A prospective observational study. J Crit Care 2018; 47:41-48. [DOI: 10.1016/j.jcrc.2018.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/28/2018] [Accepted: 06/01/2018] [Indexed: 01/18/2023]
|
18
|
Differences in IgG Fc Glycosylation Are Associated with Outcome of Pediatric Meningococcal Sepsis. mBio 2018; 9:mBio.00546-18. [PMID: 29921663 PMCID: PMC6016251 DOI: 10.1128/mbio.00546-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pediatric meningococcal sepsis often results in morbidity and/or death, especially in young children. Our understanding of the reasons why young children are more susceptible to both the meningococcal infection itself and a more fulminant course of the disease is limited. Immunoglobulin G (IgG) is involved in the adaptive immune response against meningococcal infections, and its effector functions are highly influenced by the glycan structure attached to the fragment crystallizable (Fc) region. It was hypothesized that IgG Fc glycosylation might be related to the susceptibility and severity of meningococcal sepsis. Because of this, the differences in IgG Fc glycosylation between 60 pediatric meningococcal sepsis patients admitted to the pediatric intensive care unit and 46 age-matched healthy controls were investigated, employing liquid chromatography with mass spectrometric detection of tryptic IgG glycopeptides. In addition, Fc glycosylation profiles were compared between patients with a severe outcome (death or the need for amputation) and a nonsevere outcome. Meningococcal sepsis patients under the age of 4 years showed lower IgG1 fucosylation and higher IgG1 bisection than age-matched healthy controls. This might be a direct effect of the disease; however, it can also be a reflection of previous immunologic challenges and/or a higher susceptibility of these children to develop meningococcal sepsis. Within the young patient group, levels of IgG1 hybrid-type glycans and IgG2/3 sialylation per galactose were associated with illness severity and severe outcome. Future studies in larger groups should explore whether IgG Fc glycosylation could be a reliable predictor for meningococcal sepsis outcome. Meningococcal sepsis causes significant mortality and morbidity worldwide, especially in young children. Identification of risk factors for a more fulminant infection would help to decide on appropriate treatment strategies for the individual patients. Immunoglobulin G (IgG) plays an essential role in humoral immune responses and is involved in the adaptive immune response against meningococcal infections. Of great influence on the receptor affinity of IgG is the N-glycan on its fragment crystallizable (Fc) portion. In the present study, we analyzed IgG glycosylation during the fast development of meningococcal sepsis in children, and we were able to identify glycosylation features that are different between meningococcal sepsis patients and healthy controls. These features might be indicative of a higher susceptibility to meningococcal sepsis. In addition, we found glycosylation features in the patients that were associated with illness severity and severe disease outcome, having the potential to serve as a disease outcome predictor.
Collapse
|
19
|
Abstract
OBJECTIVES Children with meningococcal sepsis are highly at risk for fulminant disease, multiple organ failure, and death. Recently, neutrophil extracellular traps levels have been indicated as a marker for severity in different kinds of sepsis. Our aim was to study the role of neutrophil extracellular traposis in meninogococcal sepsis in children. DESIGN We measured myeloperoxidase-DNA, a marker for neutrophil extracellular traps, in serum of meningococcal sepsis patients upon admission to PICU, at 24 hours, and at 1 month and studied the association with clinical outcome. Subsequently, we tested whether Neisseria meningitidis, isolated from children with meningococcal sepsis, were able to induce neutrophil extracellular traposis, using confocal microscopy live imaging. SETTING We used enzyme-linked immunosorbent assays to measure myeloperoxidase-DNA in patient serum. We also included inflammatory markers that were previously measured in this group. PATIENTS We included exclusively children with meningococcal sepsis. INTERVENTIONS From each patient, serum was collected for analysis. MEASUREMENTS AND MAIN RESULTS Myeloperoxidase-DNA levels at admission (n = 35; median, 0.21 AU/mL; interquartile range, 0.12-0.27) and at 24 hours (n = 39; median, 0.14 AU/mL; interquartile range, 0.09-0.25) were significantly higher than the myeloperoxidase-DNA levels after 1 month (controls: n = 36; median, 0.07 AU/mL; interquartile range, 0.05-0.09; p < 0.001). We did not observe a correlation between myeloperoxidase-DNA levels and mortality, cell-free DNA, or other inflammatory markers. In addition, N. meningitidis are fast and strong inducers of neutrophil extracellular traposis. CONCLUSIONS Children admitted to PICU for meningococcal sepsis have higher neutrophil extracellular traps levels at admission and after 24 hours than controls. Neutrophil extracellular traps levels were not associated with outcome, cell-free DNA, or other inflammatory markers. These neutrophil extracellular traps may be induced by N. meningitidis, since these are strong neutrophil extracellular traposis inducers.
Collapse
|
20
|
Lécuyer H, Virion Z, Barnier JP, Matczak S, Bourdoulous S, Bianchini E, Saller F, Borgel D, Nassif X, Coureuil M. An ADAM-10 dependent EPCR shedding links meningococcal interaction with endothelial cells to purpura fulminans. PLoS Pathog 2018; 14:e1006981. [PMID: 29630665 PMCID: PMC5908201 DOI: 10.1371/journal.ppat.1006981] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 04/19/2018] [Accepted: 03/20/2018] [Indexed: 01/03/2023] Open
Abstract
Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans. Neisseria meningitidis (meningococcus) is responsible for a severe syndrome called purpura fulminans in which the coagulation system is totally dysregulated, leading to an extensive occlusion of blood microvessels. The pathogenesis of this syndrome is still not understood. Here we show that the meningococcus, when adhering on the apical surface of endothelial cells, induces the activation of membranous protease named ADAM-10, which in turn hydrolyses a cellular receptor called EPCR. The latter is key for the activation of a circulating potent anticoagulant, the Protein C (PC). PC activation is then impaired following meningococcal adhesion on endothelial cells. This work unveils for the first time a specific dysregulation of coagulation induced by the meningococcus and potentially identifies new therapeutic targets for meningococcal purpura fulminans.
Collapse
Affiliation(s)
- Hervé Lécuyer
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR8253, Paris, France
- Université Paris Descartes, Paris, France
- Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Necker Enfants Malades, Service de Microbiologie Clinique, Paris, France
- * E-mail:
| | - Zoé Virion
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR8253, Paris, France
- Université Paris Descartes, Paris, France
| | - Jean-Philippe Barnier
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR8253, Paris, France
- Université Paris Descartes, Paris, France
- Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Necker Enfants Malades, Service de Microbiologie Clinique, Paris, France
| | - Soraya Matczak
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR8253, Paris, France
- Université Paris Descartes, Paris, France
| | - Sandrine Bourdoulous
- Université Paris Descartes, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
| | - Elsa Bianchini
- INSERM UMR-S1176, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - François Saller
- INSERM UMR-S1176, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Delphine Borgel
- INSERM UMR-S1176, Université Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Necker Enfants Malades, Service d’Hématologie Biologique, Paris, France
| | - Xavier Nassif
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR8253, Paris, France
- Université Paris Descartes, Paris, France
- Assistance Publique–Hôpitaux de Paris, Hôpital Universitaire Necker Enfants Malades, Service de Microbiologie Clinique, Paris, France
| | - Mathieu Coureuil
- Institut Necker Enfants Malades, INSERM U1151, CNRS UMR8253, Paris, France
- Université Paris Descartes, Paris, France
| |
Collapse
|
21
|
Wollina U, Koch A, Heinig B, Tchernev G, Lotti T. Cutaneous Microembolism of Fingers and Toes. Open Access Maced J Med Sci 2018; 6:166-169. [PMID: 29484019 PMCID: PMC5816294 DOI: 10.3889/oamjms.2018.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 08/19/2017] [Accepted: 08/20/2017] [Indexed: 01/08/2023] Open
Abstract
A macro vascular embolism is a well-known emergency. In contrast, cutaneous microembolism is a lesser known symptom. However, cutaneous microembolism of fingers and toes is a red flag symptom for vascular emergencies. The underlying cause may involve infectious, immunological, metabolic and physical disorders, coagulation disorders and malignancies. Early recognition can help to live safe.
Collapse
Affiliation(s)
- Uwe Wollina
- Department of Dermatology and Allergology, Städtisches Klinikum Dresden, Friedrichstrasse 41, 01067 Dresden, Germany
| | - André Koch
- Department of Dermatology and Allergology, Städtisches Klinikum Dresden, Friedrichstrasse 41, 01067 Dresden, Germany
| | - Birgit Heinig
- Center for Physical Therapy and Rehabilitative Medicine, Städtisches Klinikum Dresden, Dresden, Germany
| | - Georgi Tchernev
- Department of Dermatology, Venereology and Dermatologic Surgery, Medical Institute of Ministry of Interior, Sofia, Bulgaria.,Onkoderma Policlinic for Dermatology and Dermatologic Surgery, Sofia, Bulgaria
| | | |
Collapse
|
22
|
Hematologic Manifestations of Childhood Illness. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00152-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
23
|
Stephan F, Bulder I, Luken BM, Hazelzet J, Wuillemin WA, Zeerleder S. Complexes of factor VII-activating protease with plasminogen activator inhibitor-1 in human sepsis. Thromb Haemost 2017; 112:219-21. [DOI: 10.1160/th13-12-1062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 02/14/2014] [Indexed: 01/28/2023]
|
24
|
Colling ME, Bendapudi PK. Purpura Fulminans: Mechanism and Management of Dysregulated Hemostasis. Transfus Med Rev 2017; 32:69-76. [PMID: 29157918 DOI: 10.1016/j.tmrv.2017.10.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/21/2017] [Accepted: 10/13/2017] [Indexed: 01/30/2023]
Abstract
Purpura fulminans (PF) is a highly thrombotic subtype of disseminated intravascular coagulation that can accompany severe bacterial, and more rarely, viral infections. PF is associated with an extremely high mortality rate, and patients often die of overwhelming multisystemic thrombosis rather than septic shock. Survivors typically experience amputation of involved extremities and significant scarring in affected areas. Despite the devastating clinical course associated with this hemostatic complication of infection, the mechanism of PF remains poorly understood. Severe acquired deficiency of protein C and dysfunction of the protein C-thrombomodulin pathway as well as other systems that exert a negative regulatory effect on coagulation have been implicated. Management of PF involves treatment of the underlying infection, aggressive anticoagulation, and robust transfusion support aimed at correcting acquired deficiencies in natural anticoagulant proteins. In this review, we address the diagnosis and management of PF with a focus on a rational approach to this condition informed by the available data. Proposed mechanisms underlying the dysregulation of coagulation seen in PF are also covered, and implications for therapy are discussed.
Collapse
Affiliation(s)
- Meaghan E Colling
- Department of Medicine, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Pavan K Bendapudi
- Harvard Medical School, Boston, MA; Division of Hematology, Massachusetts General Hospital, Boston, MA; Blood Transfusion Service, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
25
|
Pierce RW, Giuliano JS, Pober JS. Endothelial Cell Function and Dysfunction in Critically Ill Children. Pediatrics 2017; 140:peds.2017-0355. [PMID: 28759412 PMCID: PMC9923607 DOI: 10.1542/peds.2017-0355] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2017] [Indexed: 11/24/2022] Open
Abstract
Endothelial cells (ECs) line the lumen of the entire vascular system and actively regulate blood flow; maintain blood fluidity; control water, solute, and macromolecular transfer between blood and tissue; and modulate circulating immune cell recruitment and activation. These vital functions, combined with the broad anatomic distribution of ECs, implicate them in all forms of critical illness. The present article discusses how ECs adapt and break down during the course of critical illness. We first review the biology of ECs, highlighting the vascular segmental differences and their specific roles in the maintenance of homeostasis. We then discuss how ECs acquire new functions to restore local and systemic homeostasis (activation) as well as how breakdowns in EC functions (dysfunction) contribute to local and systemic pathologic responses, with clinical correlations. Lastly, how these processes have been studied in critically ill children is discussed.
Collapse
Affiliation(s)
- Richard W. Pierce
- Departments of Pediatrics and,Address correspondence to Richard W. Pierce, MD, MS, Department of Pediatrics, Section of Critical Care Medicine, Yale University, 333 Cedar St, PO Box 208064, New Haven, CT 06520. E-mail:
| | | | | |
Collapse
|
26
|
|
27
|
Abstract
OBJECTIVE To evaluate all published pediatric randomized controlled trials of patients with septic shock from any cause to examine the outcome measures used, the strengths and limitations of these measurements and whether the trial outcomes met feasibility criteria. DATA SOURCES We used a previously published database of pediatric critical care randomized controlled trials (PICUtrials.net) derived from searches of MEDLINE, EMBASE, LILACS, and CENTRAL. STUDY SELECTION We included randomized controlled trials of interventions to children admitted to a PICU with septic or dengue hemorrhagic shock which were published in English. DATA EXTRACTION Study characteristics and outcomes were retrieved by two independent reviewers with disagreement being resolved by a third reviewer. We defined feasibility as 1) recruitment of at least 90% of the targeted sample size and agreement of the observed outcome rate in the control group with the rate used for the sample size calculation to within 10% or 2) finding of a statistically significant difference in an interim or final analysis. DATA SYNTHESIS Nineteen of 321 identified articles were selected for review. Fourteen of 19 studies (74%) provided an a priori definition of their primary outcome measure in their "Methods section." Mortality rate was the most commonly reported primary outcome (8/14; 57%), followed by duration of shock (4/14; 29%) followed by organ failure (1/14; 7%). Only three of 19 included trials met feasibility criteria. CONCLUSIONS Our review found that use of mortality alone as a primary outcome in pediatric septic shock trials was associated with significant limitations and that long-term patient-centered outcomes were not used in this setting. Composite outcomes incorporating mortality and long-term outcomes should be explored for use in future pediatric septic shock trials.
Collapse
|
28
|
Boeddha NP, Driessen GJ, Cnossen MH, Hazelzet JA, Emonts M. Circadian Variation of Plasminogen-Activator-Inhibitor-1 Levels in Children with Meningococcal Sepsis. PLoS One 2016; 11:e0167004. [PMID: 27893784 PMCID: PMC5125643 DOI: 10.1371/journal.pone.0167004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/07/2016] [Indexed: 11/18/2022] Open
Abstract
Objective To study whether the circadian variation of plasminogen-activator-inhibitor-1 (PAI-1) levels, with high morning levels, is associated with poor outcome of children with meningococcal sepsis presenting in the morning hours. Design Retrospective analysis of prospectively collected clinical and laboratory data. Setting Single center study at Erasmus MC-Sophia Children’s Hospital, Rotterdam, the Netherlands. Subjects 184 patients aged 3 weeks to 18 years with meningococcal sepsis. In 36 of these children, PAI-1 levels at admission to the PICU were measured in plasma by ELISA. Interventions None. Measurements and main results Circadian variation was studied by dividing one day in blocks of 6 hours. Patients admitted between 6:00 am and 12:00 am had increased illness severity scores and higher PAI-1 levels (n = 9, median 6912 ng/mL, IQR 5808–15600) compared to patients admitted at night (P = 0.019, n = 9, median 3546 ng/mL, IQR 1668–6118) or in the afternoon (P = 0.007, n = 7, median 4224 ng/mL, IQR 1804–5790). In 184 patients, analysis of circadian variation in relation to outcome showed more deaths, amputations and need for skin grafts in patients admitted to the PICU between 6:00 am and 12:00 am than patients admitted during the rest of the day (P = 0.009). Conclusions Circadian variation of PAI-1 levels is present in children with meningococcal sepsis and is associated with illness severity, with a peak level in the morning. Whether circadian variation is an independent risk factor for morbidity and mortality in meningococcal sepsis needs to be explored in future studies.
Collapse
Affiliation(s)
- Navin P. Boeddha
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Division of Pediatric Infectious Diseases & Immunology, Erasmus MC-Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gertjan J. Driessen
- Department of Pediatrics, Division of Pediatric Infectious Diseases & Immunology, Erasmus MC-Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marjon H. Cnossen
- Department of Pediatrics, Division of Pediatric Hematology, Erasmus MC-Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jan A. Hazelzet
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- * E-mail:
| | - Marieke Emonts
- Paediatric Infectious Diseases and Immunology Department, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
29
|
Fatal purpura fulminans and Waterhouse-Friderichsen syndrome from fulminant Streptococcus pneumoniae sepsis in an asplenic young adult. IDCases 2016; 6:1-4. [PMID: 27583208 PMCID: PMC4995527 DOI: 10.1016/j.idcr.2016.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/10/2016] [Accepted: 08/10/2016] [Indexed: 11/23/2022] Open
Abstract
Asplenic patients are at increased risk for sepsis and fulminant infection. Sepsis in these patients is typically secondary to encapsulated bacteria, with Streptococcus pneumoniae being the most frequent pathogen. Rare complications of severe sepsis include purpura fulminans and bilateral adrenal hemorrhage (Waterhouse-Friderichsen syndrome). We present the case of a 36-year-old woman, healthy except for splenectomy years prior for idiopathic thrombocytopenic purpura treatment, who presented with fever. Upon presentation to our hospital, three hours after symptoms onset, she had purpura fulminans and shock. Despite timely antimicrobials and maximal resuscitative efforts, her disease progressed and she expired 12 hours after symptoms onset. Autopsy revealed bilateral adrenal hemorrhage; acute adrenal crisis likely contributed to her refractory shock. Prior to her presentation, she had not received guideline-based post-splenectomy care. Sepsis in asplenic patients can be fulminant and rapidly fatal. Streptococcus pneumoniae remains the most frequent cause, despite decreasing rates in recent years related to widespread pneumococcal vaccination. Guideline-based vaccinations and "pill-in-pocket" therapy can be life-saving for asplenic patients. Purpura fulminans represents an extreme manifestation of disseminated intravascular coagulation, is more common in asplenic patients, and portends a poor prognosis. Waterhouse-Friderichsen syndrome can be seen concurrently with purpura fulminans and further portends a poor prognosis; pre-mortem diagnosis requires a high index of suspicion.
Collapse
|
30
|
Pappalardo F, Crivellari M, Di Prima AL, Agracheva N, Celinska-Spodar M, Lembo R, Taddeo D, Landoni G, Zangrillo A. Protein C zymogen in severe sepsis: a double-blinded, placebo-controlled, randomized study. Intensive Care Med 2016; 42:1706-1714. [PMID: 27344436 DOI: 10.1007/s00134-016-4405-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/23/2016] [Indexed: 12/22/2022]
Abstract
PURPOSE To determine whether protein C zymogen (protein C concentrates or human protein C) improves clinically relevant outcomes in adult patients with severe sepsis and septic shock. METHODS This is a randomized, double-blind, placebo-controlled, parallel-group trial that from September 2012 to June 2014 enrolled adult patients with severe sepsis or septic shock and high risk of death and of bleeding (e.g., APACHE II greater than 25, extracorporeal membrane oxygenation or disseminated intravascular coagulopathy). All patients completed their follow-up 90 days after randomization and data were analyzed according to the intention-to-treat principle. Follow-up was performed at 30 and 90 days after randomization. The primary endpoint was a composite outcome of prolonged intensive care unit (ICU) stay and/or 30-day mortality. Secondary endpoints included mortality. RESULTS The study was stopped early in a situation of futility for the composite outcome of prolonged ICU stay and/or 30-day mortality that was 79 % (15 patients) in the protein C zymogen group and 67 % (12 patients) in the placebo group (p = 0.40) and for a concomitant safety issue: ICU mortality was 79 % (15 patients) in the protein C zymogen group vs 39 % (7 patients) in the placebo group (p = 0.020), and 30-day mortality was 68 vs 39 % (p = 0.072). CONCLUSION Protein C zymogen did not improve clinically relevant outcomes in severe sepsis and septic shock adult patients. Given its high cost and the potential increase in mortality, the use of this drug in adult patients should be discouraged.
Collapse
Affiliation(s)
- Federico Pappalardo
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Martina Crivellari
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Ambra L Di Prima
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Nataliya Agracheva
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Städtiches Klinikum Braunschweigh, Braunschweig, Germany
| | - Malgorzata Celinska-Spodar
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Institute of Cardiology, Warsaw, Poland
| | - Rosalba Lembo
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Daiana Taddeo
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Ospedale di Roccadaspide, Salerno, Italy
| | - Giovanni Landoni
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alberto Zangrillo
- IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
31
|
Perillo T, Muggeo P, Arcamone G, Leonardis FD, Santoro N. Non Activated Protein C Supplementation in Septic Pediatric Hematological Patients. Pediatr Rep 2016; 8:6488. [PMID: 27433305 PMCID: PMC4933811 DOI: 10.4081/pr.2016.6488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 04/17/2016] [Accepted: 05/23/2016] [Indexed: 12/29/2022] Open
Abstract
The purpose of the study was to examine safety and efficacy of non-activated Protein C (PC) supplementation in our cohort of septic pediatric hematological patients. We conducted a retrospective study of 22 septic patients receiving human plasma-derived PC concentrate from 2008 to 2015 at our Pediatric Oncology Center (Bari, Italy). The Surviving sepsis campaign definitions for sepsis, severe sepsis and septic shock were used to define the patients' septic status. For each patient, we calculated Lansky performance status scale (LPSS) and a risk score defined the Hematologic risk score (HRS) that we created in 2007. Patients were defined as High risk for severe sepsis/septic shock in case of HRS>3. HRS<3 identified low risk patients. Baseline serum PC levels, PC administration dosage and duration and days until a 20% improvement in LPSS. Observed baseline serum PC levels (bPC) blood concentrations ranged from 31 to 80%. Patients received PC supplementation in case of low age-related bPC levels or >10% PC concentration decrease within 12 hours from the first evaluation. All patients received 80 U/kg/day PC, intravenously, every twenty-four hours. No drug-related adverse event was observed. The observed sepsis-related mortality rate in our cohort was 9%. PC supplementation in our cohort appeared to be safe, and, probably due to prompt PC administration, we observed an overall mortality that was much lower than expected mortality in cancer severe septic patients.
Collapse
Affiliation(s)
- Teresa Perillo
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Bari , Italy
| | - Paola Muggeo
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Bari , Italy
| | - Giampaolo Arcamone
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Bari , Italy
| | - Francesco De Leonardis
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Bari , Italy
| | - Nicola Santoro
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Bari , Italy
| |
Collapse
|
32
|
Abstract
Thrombocytopenia-associated multiple organ failure (TAMOF) is a clinical phenotype that encompasses a spectrum of syndromes associated with disseminated microvascular thromboses, such as the thrombotic microangiopathies thrombotic thrombocytopenic purpura/hemolytic uremic syndrome (TTP/HUS) and disseminated intravascular coagulation (DIC). Autopsies findings in TTP, HUS, or DIC reveal specific findings that can differentiate these 3 entities. Von Willebrand factor and ADAMTS-13 play a central role in TTP. Shiga toxins and the complement pathway are vital in the development of HUS. Tissue factor is the major protease that drives the pathology of DIC. Acute kidney injury (AKI) is a common feature in patients with TAMOF.
Collapse
|
33
|
Nguyen TC, Gushiken F, Correa JI, Dong JF, Dasgupta SK, Thiagarajan P, Cruz MA. A recombinant fragment of von Willebrand factor reduces fibrin-rich microthrombi formation in mice with endotoxemia. Thromb Res 2015; 135:1025-30. [PMID: 25769494 DOI: 10.1016/j.thromres.2015.02.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Disseminated fibrin deposition in the microvasculature such as in disseminated intravascular coagulation (DIC) arises from uninhibited activated coagulation secondary to sustained systemic inflammation. Currently there is no treatment for DIC. Treating the underlying trigger and supportive care are the current recommendations to manage DIC. This study aims at using recombinant von Willebrand factor (VWF) A2 domain polypeptide to inhibit VWF-mediated platelet adhesion to fibrin and prevent DIC. MATERIALS AND METHODS We use flow chamber assay to test the capacity of purified A2 protein to inhibit platelet adhesion to immobilized fibrin(ogen) and platelet-fibrin clot formation. We use a murine model of lipopolysaccharide-induced DIC to examine the effect of A2 protein on DIC. RESULTS The A2 protein blocked flow-dependent platelet adhesion to fibrin, delayed fibrin polymerization, and inhibited platelet-fibrin clot formation in vitro. The infusion of the purified A2 protein to the endotoxin-treated mice prevented fibrin-rich microthrombi formation in brain, lung, kidney, and liver. It also attenuated levels of inflammatory mediators, and markedly reduced mortality rates at 96hours. CONCLUSIONS The A2 protein inhibited platelet interaction with fibrin(ogen). Furthermore, A2 prevented disseminated fibrin-rich microthrombi and decrease mortality in a lipopolysaccharide-induced DIC murine model. A2 could provide a novel therapeutic approach in critically ill patients with uninhibited activated coagulation and disseminated fibrin deposition such as DIC.
Collapse
Affiliation(s)
- Trung C Nguyen
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, TX 77030; Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX 77030; Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States
| | - Francisca Gushiken
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States
| | - Juliana I Correa
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Jing-Fei Dong
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Swapan K Dasgupta
- Department of Pathology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States; Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States
| | - Perumal Thiagarajan
- Department of Pathology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States; Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States
| | - Miguel A Cruz
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX 77030; Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States.
| |
Collapse
|
34
|
Brunkhorst FM, Patchev V. [Sepsis-associated Purpura Fulminans International Registry--Europe (SAPFIRE)]. Med Klin Intensivmed Notfmed 2014; 109:591-5. [PMID: 25348051 DOI: 10.1007/s00063-014-0402-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 09/16/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Purpura fulminans is a rare life-threatening condition which is characterized by disseminated thrombosis in dermal and systemic microcirculation, cutaneous hemorrhages with progressing necrosis and multiple organ failure. The underlying pathogenesis is based on the disruption of the intrinsic anticoagulation cascade, with protein C deficiency being considered the leading factor in this process. In the majority of cases, the condition emerges as consumptive coagulopathy associated with severe sepsis. OBJECTIVES Epidemiological data on sepsis-associated purpura fulminans (SAPF) are scarce and evidence-based treatment guidelines have not been established yet. While restoration of the balance in the coagulation cascade is a declared therapeutic goal, evaluations of the efficacy of different therapeutic approaches in randomized clinical trials are still lacking. The causal role of individual microbial pathogens also requires comprehensive evaluation. METHODS A prospective multicenter Sepsis-Associated Purpura Fulminans International Registry-Europe (SAPFIRE) will be established in the first quarter of 2015. For the first time, participating centers will systematically collect information on etiology, clinical course, biomarkers, treatment, morbidity, and mortality of SAPF. RESULTS The SAPFIRE data will be periodically evaluated and disseminated. Retrospective analysis of each center's data and regular access to aggregated information collected by other centers will enable the participants to monitor and update care quality standards.
Collapse
Affiliation(s)
- F M Brunkhorst
- Center of Sepsis Control and Care (CSCC), Klinik für Anaesthesiologie und Intensivtherapie, Universitätsklinikum Jena, Salvador-Allende-Platz 29, 07747, Jena, Deutschland,
| | | |
Collapse
|
35
|
Piccin A, O' Marcaigh A, Mc Mahon C, Murphy C, Okafor I, Marcheselli L, Casey W, Claffey L, Smith OP. Non-activated plasma-derived PC improves amputation rate of children undergoing sepsis. Thromb Res 2014; 134:63-7. [PMID: 24821370 DOI: 10.1016/j.thromres.2014.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/11/2014] [Accepted: 04/17/2014] [Indexed: 11/24/2022]
Abstract
Low circulating protein C (PC) levels have been observed in sepsis, especially in patients with Neisseriae Meningitides infections. Poor clinical outcome and high limb amputation rates have been associated in infected patients with low circulating PC levels. Published studies using activated PC replacement therapy patients with sepsis have shown reduced mortality rates, however, its use has been associated with severe bleeding events. Paediatric sepsis studies using non-activated plasma-derived PC (Ceprotin®) are lacking. We present a retrospective study in children with sepsis who were treated with Ceprotin® focusing on amputation rate post treatment. Thirty subjects were identified. Median age at diagnosis was 2 years. Twenty-one (70%) were treated for Nesseria Meningitides and one (3%) for Streptococcus-A β-haemolyticus, another 8 (26%) patients with malignancies were treated for neutropenic sepsis. Following Ceprotin® administration, a significant increase in leukocyte count (p=0.004), neutrophil count (p=0.001) and PC (pretreatment=13%, posttreatment=88.5%; p=0.0001) was seen. Prothrombin time (pretreatment =30.3 seconds, posttreatment =16.5; p=0.000) and activated partial thromboplastin time (pretreatment =61.8 sec, postreatment =42.6 sec; p=0.000) were significantly reduced, while fibrinogen levels were significantly elevated (pretreatment =1.9 g/dL, posttreatment =4.4 g/dL; p=0.000). The median time between admission to intensive care and Ceprotin® administration was 10 hrs. Limb amputation rate was reduced (16-23% versus 30-50% from previous studies) and there were no haemorrhagic events observed. This study demonstrates the safe administration of non-activated plasma-derived PC concentrate in patients with sepsis who are coagulopathic and it associated with a reduction in amputation rates.
Collapse
Affiliation(s)
- Andrea Piccin
- Hematology Dept, Our Lady's Children's Hospital, Dublin, Ireland; Hematology Dept, San Maurizio Regional Hospital, Bolzano, Italy; Trinity College University, Dublin, Ireland.
| | | | - Corrina Mc Mahon
- Hematology Dept, Our Lady's Children's Hospital, Dublin, Ireland
| | | | - Ikechukwu Okafor
- Hematology Dept, Our Lady's Children's Hospital, Dublin, Ireland
| | - Luigi Marcheselli
- Department of Diagnostic and Clinical Medicine and Public Health University of Modena and Reggio Emilia, Modena Italy
| | - William Casey
- Anesthesiology Dept, Our Lady's Children's Hospital, Dublin, Ireland
| | - Liam Claffey
- Anesthesiology Dept, Temple Street Hospital, Dublin, Ireland
| | - Owen Patrick Smith
- Hematology Dept, Our Lady's Children's Hospital, Dublin, Ireland; Trinity College University, Dublin, Ireland
| |
Collapse
|
36
|
|
37
|
Kager LM, Weehuizen TA, Wiersinga WJ, Roelofs JJTH, Meijers JCM, Dondorp AM, van 't Veer C, van der Poll T. Endogenous α2-antiplasmin is protective during severe gram-negative sepsis (melioidosis). Am J Respir Crit Care Med 2013; 188:967-75. [PMID: 23992406 DOI: 10.1164/rccm.201307-1344oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE α2-Antiplasmin (A2AP) is a major inhibitor of fibrinolysis by virtue of its capacity to inhibit plasmin. Although the fibrinolytic system is strongly affected by infection, the functional role of A2AP in the host response to sepsis is unknown. OBJECTIVES To study the role of A2AP in melioidosis, a common form of community-acquired sepsis in Southeast Asia and Northern Australia caused by the gram-negative bacterium Burkholderia pseudomallei. METHODS In a single-center observational study A2AP was measured in patients with culture-proven septic melioidosis. Wild-type and A2AP-deficient (A2AP(-/-)) mice were intranasally infected with B. pseudomallei to induce severe pneumosepsis (melioidosis). Parameters of inflammation and coagulation were measured, and survival studies were performed. MEASUREMENTS AND MAIN RESULTS Patients with melioidosis showed elevated A2AP plasma levels. Likewise, A2AP levels in plasma and lung homogenates were elevated in mice infected with B. pseudomallei. A2AP-deficient (A2AP(-/-)) mice had a strongly disturbed host response during experimental melioidosis as reflected by enhanced bacterial growth at the primary site of infection accompanied by increased dissemination to distant organs. In addition, A2AP(-/-) mice showed more severe lung pathology and injury together with an increased accumulation of neutrophils and higher cytokine levels in lung tissue. A2AP deficiency further was associated with exaggerated systemic inflammation and coagulation, increased distant organ injury, and enhanced lethality. CONCLUSIONS This study is the first to identify A2AP as a protective mediator during gram-negative (pneumo)sepsis by limiting bacterial growth, inflammation, tissue injury, and coagulation.
Collapse
|
38
|
Campsall PA, Laupland KB, Niven DJ. Severe meningococcal infection: a review of epidemiology, diagnosis, and management. Crit Care Clin 2013; 29:393-409. [PMID: 23830646 DOI: 10.1016/j.ccc.2013.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neisseria meningitidis, also known as meningococcus, is a relatively uncommon cause of invasive infection, but when it occurs it is frequently severe and potentially life threatening. Meningococcus should be considered and investigated promptly as a potentially etiologic pathogen in any patient with meningitis, or sepsis accompanied by a petechial rash. Suspected patients should receive early appropriate antimicrobial therapy concomitantly with confirmatory invasive diagnostic tests. Vaccines have reduced the incidence of infection with certain non-B meningococcal serogroups, and new serotype B vaccines are on the horizon. This article reviews the epidemiology, diagnosis, and management of severe meningococcal infections.
Collapse
Affiliation(s)
- Paul A Campsall
- Department of Critical Care Medicine, University of Calgary and Alberta Health Services, 3500 26th Avenue Northeast, Calgary, Alberta T1Y 6J4, Canada
| | | | | |
Collapse
|
39
|
Abstract
Bleeding in patients in pediatric intensive care units is associated with an increased risk of mortality. Fortunately, most patients with an abnormal coagulation profile do not bleed because this is generally secondary to liver disease or dietary-induced vitamin K deficiency. When the laboratory markers of coagulopathy are the result of disseminated intravascular coagulation, bleeding is common and the risk of mortality extreme. Although interventions directed toward correcting the abnormal coagulation test results are generally initiated, they are also generally either not warranted or not fully successful.
Collapse
Affiliation(s)
- Robert I Parker
- Pediatric Hematology/Oncology, Stony Brook Long Island Children's Hospital, Stony Brook University School of Medicine, 100 Nicolls Road, Stony Brook, NY 11794, USA.
| |
Collapse
|
40
|
Decreased expression of serum and microvascular vascular endothelial growth factor receptor-2 in meningococcal sepsis*. Pediatr Crit Care Med 2013; 14:682-5. [PMID: 23842590 DOI: 10.1097/pcc.0b013e3182917ccb] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To determine the skin microvessel expression of vascular endothelial growth factor receptor 2 and serum-soluble vascular endothelial growth factor receptor 2 levels in children with meningococcal sepsis. DESIGN Observational study. SETTING Two tertiary academic children hospital PICUs. PATIENTS Children with meningococcal sepsis. INTERVENTION Skin biopsy and blood sample collection. MEASUREMENTS AND MAIN RESULTS Determination of skin microvessel vascular endothelial growth factor receptor 2 expression in skin biopsies by immunohistochemistry and measurement of serum-soluble vascular endothelial growth factor receptor 2 by enzyme-linked immunosorbent assay. Percentage of vascular endothelial growth factor receptor 2-positive skin microvessels and the staining intensity were significantly lower in children with meningococcal sepsis (n = 10) compared to controls (7.6% ± 8.8% vs 44.6% ± 39.2%; p = 0.009 and 0.7% ± 0.7% vs 1.7% ± 1.1%; p = 0.033, respectively). In addition, circulating serum levels of soluble vascular endothelial growth factor receptor 2 were decreased in sepsis (8,148 ± 1,140 pg/mL vs 13,414 ± 2,692 pg/mL; p < 0.001). Serum-soluble vascular endothelial growth factor receptor 2 levels (n = 28) were inversely correlated with Pediatric Risk of Mortality III score (r = -0.43; p = 0.023) and more decreased in nonsurvivors compared to survivors (5,640 ± 1,940 pg/mL vs 7,378 ± 2,336 pg/mL; p = 0.037). CONCLUSIONS Microvascular expression of vascular endothelial growth factor receptor 2 and serum-soluble vascular endothelial growth factor receptor 2 levels are decreased in children with sepsis. Serum-soluble vascular endothelial growth factor receptor 2 levels are inversely correlated with disease severity indicated by Pediatric Risk of Mortality III score and survival. Decreased vascular endothelial growth factor receptor 2 expression may hinder natural recovery from sepsis-associated microvascular injury and the effectiveness of therapeutic strategies targeting vascular endothelial growth factor-vascular endothelial growth factor receptor 2 signaling in sepsis patients.
Collapse
|
41
|
Christiaans SC, Wagener BM, Esmon CT, Pittet JF. Protein C and acute inflammation: a clinical and biological perspective. Am J Physiol Lung Cell Mol Physiol 2013; 305:L455-66. [PMID: 23911436 DOI: 10.1152/ajplung.00093.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The protein C system plays an active role in modulating severe systemic inflammatory processes such as sepsis, trauma, and acute respiratory distress syndrome (ARDS) via its anticoagulant and anti-inflammatory properties. Plasma levels of activated protein C (aPC) are lower than normal in acute inflammation in humans, except early after severe trauma when high plasma levels of aPC may play a mechanistic role in the development of posttraumatic coagulopathy. Thus, following positive results of preclinical studies, a clinical trial (PROWESS) with high continuous doses of recombinant human aPC given for 4 days demonstrated a survival benefit in patients with severe sepsis. This result was not confirmed by subsequent clinical trials, including the recently published PROWESS-SHOCK trial in patients with septic shock and a phase II trial with patients with nonseptic ARDS. A possible explanation for the major difference in outcome between PROWESS and PROWESS-SHOCK trials is that lung-protective ventilation was used for the patients included in the recent PROWESS-SHOCK, but not in the original PROWESS trial. Since up to 75% of sepsis originates from the lung, aPC treatment may not have added enough to the beneficial effect of lung-protective ventilation to show lower mortality. Thus whether aPC will continue to be used to modulate the acute inflammatory response in humans remains uncertain. Because recombinant human aPC has been withdrawn from the market, a better understanding of the complex interactions between coagulation and inflammation is needed before considering the development of new drugs that modulate both coagulation and acute inflammation in humans.
Collapse
Affiliation(s)
- Sarah C Christiaans
- Dept. of Anesthesiology, Univ. of Alabama at Birmingham, 619 S. 19th St., JT926, Birmingham, AL 35249.
| | | | | | | |
Collapse
|
42
|
van der Flier M, Sharma DB, Estevão S, Emonts M, Rook D, Hazelzet JA, van Goudoever JB, Hartwig NG. Increased CD4(+) T cell co-inhibitory immune receptor CEACAM1 in neonatal sepsis and soluble-CEACAM1 in meningococcal sepsis: a role in sepsis-associated immune suppression? PLoS One 2013; 8:e68294. [PMID: 23894299 PMCID: PMC3718779 DOI: 10.1371/journal.pone.0068294] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/28/2013] [Indexed: 12/16/2022] Open
Abstract
The co-inhibitory immune receptor carcinoembryonic antigen-related cell-adhesion molecule 1 (CEACAM1) and its self-ligand CEACAM1 can suppress T cell function. Suppression of T cell function in sepsis is well documented. Late-onset neonatal sepsis in VLBW-infants was associated with an increased percentage CEACAM1 positive CD4+ T-cells. Meningococcal septic shock in children was associated with increased serum soluble CEACAM1. In conclusion our data demonstrate increased surface expression of the co-inhibitory immune receptor CEACAM1 in late-onset neonatal sepsis in VLBW-infants, and increased circulating soluble CEACAM1 in children with meningococcal sepsis. Increased T-cell CEACAM1 expression and increased circulating soluble CEACAM1 may contribute to sepsis-associated immune suppression.
Collapse
Affiliation(s)
- Michiel van der Flier
- Department of Pediatric Infectious Diseases and Immunology, Erasmus MC - Sophia, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Borgel D, Lerolle N. Quel avenir pour les médicaments de l’hémostase dans le traitement du sepsis sévère après le Xigris® ? MEDECINE INTENSIVE REANIMATION 2013. [DOI: 10.1007/s13546-013-0665-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
44
|
Frawley G, Bennett M, Thistlethwaite K, Banham N. Australian paediatric hyperbaric oxygen therapy 1998-2011. Anaesth Intensive Care 2013; 41:74-81. [PMID: 23362893 DOI: 10.1177/0310057x1304100113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
For a large number of ischaemic, infective, inflammatory or traumatic conditions, hyperbaric oxygen therapy is either the only treatment or an adjunct that significantly reduces morbidity and mortality. The primary aim of this review is to identify clinical conditions treated in a paediatric population referred to Australian hyperbaric units. Secondary aims are to describe outcomes of treatment and detail any complications occurring during treatment or during transfer between units. This was a retrospective cohort study (January 1998-December 2011) of children treated at four Australian hyperbaric medical units. A total of 112 children underwent 1099 hyperbaric treatments for 14 indications. Ages were not normally distributed with a median age of 14 years (interquartile range 11-16; range 0.25-16 years). Treatments were completed as planned in 81.5% of cases with 25 patients' treatment terminated at the request of physicians, parents or patients. Complications relating to hyperbaric oxygen therapy occurred in 58 treatments (5.3%). Central nervous system oxygen toxicity occurred in 1:366 treatments. Our findings indicate that provision of hyperbaric oxygen therapy to children is feasible in major regional hyperbaric units and is associated with low complication rates. Management of children in an adult hyperbaric facility, however, requires significant cooperation between paediatric, intensive care and hyperbaric consultants, as the need for transfer to another hospital and prolonged transports often impacts on optimal ongoing surgical and intensive care management.
Collapse
Affiliation(s)
- G Frawley
- Alfred Hyperbaric Unit, Alfred Hospital, Melbourne, Victoria.
| | | | | | | |
Collapse
|
45
|
Circulating nucleosomes and severity of illness in children suffering from meningococcal sepsis treated with protein C. Crit Care Med 2012; 40:3224-9. [DOI: 10.1097/ccm.0b013e318265695f] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
46
|
Abstract
It was suggested more than 30 yrs ago that inhibition of the clotting cascade by natural anticoagulants could decrease the high mortality observed in patients suffering from severe sepsis and septic shock. Unfortunately, this therapeutic "paradigm" has led to a dead end, illustrated by the failure of all randomized trials and the recent withdrawal of recombinant activated protein C. Should we now definitely give up trying to treat septic coagulation disturbances or is there any therapeutic alternative?
Collapse
|
47
|
Biomarker response to drotrecogin alfa (activated) in children with severe sepsis: results from the RESOLVE clinical trial*. Pediatr Crit Care Med 2012; 13:639-45. [PMID: 22791090 DOI: 10.1097/pcc.0b013e318250ad48] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE REsearching severe Sepsis and Organ dysfunction in children: A gLobal perspective (RESOLVE), a phase III trial of drotrecogin alfa (activated) in pediatric severe sepsis, examined biomarker changes in inflammation and coagulation. This report describes biomarker profiles in early severe sepsis and the pharmacodynamic assessment of drotrecogin alfa (activated) in RESOLVE. DESIGN Serial measurements of interleukin-1β, interleukin-6, interleukin-8, interleukin-10, tissue necrosis factor-α, procalcitonin, D-dimer, and thrombin-antithrombin complex were performed at baseline and daily over the first five study days. Protein C levels were performed at baseline and at the end of the 96-hr study drug infusion. Analysis of variance-based log-transformed data compared the treatment groups for each measured variable. SETTING : One hundred four pediatric intensive care units in 18 countries. PATIENTS Four hundred seventy-seven children between 38 wks corrected gestational age and 17 yrs with sepsis-induced cardiovascular and respiratory dysfunction. INTERVENTIONS Drotrecogin alfa (activated). MEASUREMENTS AND MAIN RESULTS Pharmacodynamic activity of drotrecogin alfa (activated) compared with placebo was observed with reduction of D-dimer on day 1 (p < .01) and thrombin-antithrombin complex on days 1-4 (p < .05). There were no significant changes by treatment in multiple cytokines or procalcitonin. In the overall population, a median protein C difference was not observed (p > .05) with drotrecogin alfa (activated) administration compared with placebo, although a difference (median percentage change from baseline) in favor of drotrecogin alfa (activated) was observed in patients >1 yr old (p = .0449). CONCLUSIONS While children in the RESOLVE trial were similar to adults in that they showed a relationship between severity of coagulation and inflammation abnormalities and mortality, their pharmacodynamic response to drotrecogin alfa (activated) differed with respect to changes in protein C activity and systemic inflammation.
Collapse
|
48
|
Di Nisio M, Baudo F, Cosmi B, D'Angelo A, De Gasperi A, Malato A, Schiavoni M, Squizzato A. Diagnosis and treatment of disseminated intravascular coagulation: Guidelines of the Italian Society for Haemostasis and Thrombosis (SISET). Thromb Res 2012; 129:e177-84. [DOI: 10.1016/j.thromres.2011.08.028] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/21/2011] [Accepted: 08/28/2011] [Indexed: 02/08/2023]
|
49
|
Della Valle P, Pavani G, D'Angelo A. The protein C pathway and sepsis. Thromb Res 2011; 129:296-300. [PMID: 22154246 DOI: 10.1016/j.thromres.2011.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Revised: 11/07/2011] [Accepted: 11/08/2011] [Indexed: 10/14/2022]
Abstract
After the discovery of the key components of the protein C (PC) pathway a beneficial effect on survival of the infusion of activated protein C (APC) in animal models of sepsis was demonstrated, leading to the development of recombinant human activated protein C (rh-APC) as a therapeutic agent. It soon became clear that rather than the anticoagulant and profibrinolytic activities of APC, its anti-inflammatory and cytoprotective properties played a major role in the treatment of patients with severe sepsis. Such properties affect the response to inflammation of endothelial cells and leukocytes and are exerted through binding of APC to at least five receptors with intracellular signaling. The main APC protective mechanism involves binding of the Gla-domain to the endothelial protein C receptor (EPCR) and cleavage of protease activated receptor 1 (PAR-1), eliciting suppression of proinflammatory cytokines synthesis and of intracellular proapoptotic pathways and activation of endothelial barrier properties. However, thrombin cleaves PAR-1 with much higher catalytic efficiency, followed by pro-inflammatory, pro-apoptotic and barrier disruptive intracellular signaling, and it is unclear how APC can exert a protective activity through the cleavage of PAR-1 when thrombin is also present in the same environment. Interestingly, in endothelial cell cultures, PAR-1 cleavage by thrombin results in anti-inflammatory and barrier protective signaling provided occupation of EPCR by the PC gla-domain, raising the possibility that the beneficial effects of rh-APC might be recapitulated in vivo by administration of h-PC zymogen to patients with severe sepsis. Recent reports of h-PC infusion in animal models of sepsis support this hypothesis.
Collapse
Affiliation(s)
- Patrizia Della Valle
- Coagulation Service & Thrombosis Research Unit, Scientific Institute San Raffaele, Milano, Italy
| | | | | |
Collapse
|
50
|
Saracco P, Vitale P, Scolfaro C, Pollio B, Pagliarino M, Timeus F. The coagulopathy in sepsis: significance and implications for treatment. Pediatr Rep 2011; 3:e30. [PMID: 22355515 PMCID: PMC3283198 DOI: 10.4081/pr.2011.e30] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/26/2011] [Indexed: 01/19/2023] Open
Abstract
Sepsis related coagulopathy ranges from mild laboratory alterations up to severe disseminated intravascular coagulation (DIC). There is evidence that DIC is involved in the pathogenesis of microvascular dysfunction contributing to organ failure. Additionally, the systemic activation of coagulation, by consuming platelets and coagulation factors, may cause bleeding. Thrombin generation via the tissue factor/factor VIIa route, contemporary depression of antithrombin and protein C anticoagulant systems, as well as impaired fibrin degradation, due to high circulating levels of PAI-1, contribute to enhanced intravascular fibrin deposition. This deranged coagulopathy is an independent predictor of clinical outcome in patients with severe sepsis. Innovative supportive strategies aiming at the inhibition of coagulation activation should comprise inhibition of tissue factor-mediated activation or restoration of physiological anticoagulant pathways, as the administration of recombinant human activated protein C or concentrate. In spite of some promising initial studies, additional trials are needed to define their clinical effectiveness in adults and children with severe sepsis.
Collapse
Affiliation(s)
- Paola Saracco
- Hematology Unit, Department of Pediatrics, University of Turin
| | | | | | | | | | | |
Collapse
|