1
|
Lin L, Liu H, Zhang D, Du L, Zhang H. Nanolevel Immunomodulators in Sepsis: Novel Roles, Current Perspectives, and Future Directions. Int J Nanomedicine 2024; 19:12529-12556. [PMID: 39606559 PMCID: PMC11600945 DOI: 10.2147/ijn.s496456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Sepsis represents a profound challenge in critical care, characterized by a severe systemic inflammatory response which can lead to multi-organ failure and death. The intricate pathophysiology of sepsis involves an overwhelming immune reaction that disrupts normal host defense mechanisms, necessitating innovative approaches to modulation. Nanoscale immunomodulators, with their precision targeting and controlled release capabilities, have emerged as a potent solution to recalibrate immune responses in sepsis. This review explores the recent advancements in nanotechnology for sepsis management, emphasizing the integration of nanoparticulate systems to modulate immune function and inflammatory pathways. Discussions detail the development of the immune system, the distinct inflammatory responses triggered by sepsis, and the scientific principles underpinning nanoscale immunomodulation, including specific targeting mechanisms and delivery systems. The review highlights nanoformulation designs aimed at enhancing bioavailability, stability, and therapeutic efficacy, which shows promise in clinical settings by modulating key inflammatory pathways. Ultimately, this review synthesizes the current state of knowledge and projects future directions for research, underscoring the transformative potential of nanolevel immunomodulators for sepsis treatment through innovative technologies and therapeutic strategies.
Collapse
Affiliation(s)
- Liangkang Lin
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Hanyou Liu
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Dingshan Zhang
- Department of Intensive Care Unit, Public Health Clinical Center of Chengdu, Chengdu, People’s Republic of China
| | - Lijia Du
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
2
|
Noone D, Preston RJS, Rehill AM. The Role of Myeloid Cells in Thromboinflammatory Disease. Semin Thromb Hemost 2024; 50:998-1011. [PMID: 38547918 DOI: 10.1055/s-0044-1782660] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor-mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell-mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
Collapse
Affiliation(s)
- David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
3
|
Kim H, Son B, Seo EU, Kwon M, Ahn JH, Shin H, Song GY, Park EJ, Na DH, Cho S, Kim HN, Park HH, Lee W. Cleavage-Responsive Biofactory T Cells Suppress Infectious Diseases-Associated Hypercytokinemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201883. [PMID: 35751470 PMCID: PMC9475519 DOI: 10.1002/advs.202201883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/06/2022] [Indexed: 06/15/2023]
Abstract
Severe infectious diseases, such as coronavirus disease 2019 (COVID-19), can induce hypercytokinemia and multiple organ failure. In spite of the growing demand for peptide therapeutics against infectious diseases, current small molecule-based strategies still require frequent administration due to limited half-life and enzymatic digestion in blood. To overcome this challenge, a strategy to continuously express multi-level therapeutic peptide drugs on the surface of immune cells, is established. Here, chimeric T cells stably expressing therapeutic peptides are presented for treatment of severe infectious diseases. Using lentiviral system, T cells are engineered to express multi-level therapeutic peptides with matrix metallopeptidases- (MMP-) and tumor necrosis factor alpha converting enzyme- (TACE-) responsive cleavage sites on the surface. The enzymatic cleavage releases γ-carboxyglutamic acid of protein C (PC-Gla) domain and thrombin receptor agonist peptide (TRAP), which activate endothelial protein C receptor (EPCR) and protease-activated receptor-1 (PAR-1), respectively. These chimeric T cells prevent vascular damage in tissue-engineered blood vessel and suppress hypercytokinemia and lung tissue damages in vivo, demonstrating promise for use of engineered T cells against sepsis and other infectious-related diseases.
Collapse
Affiliation(s)
- Hyelim Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Department of BiotechnologyYonsei UniversitySeoul03722Republic of Korea
| | - Boram Son
- Department of BioengineeringHanyang UniversitySeoul04763Republic of Korea
| | - Eun U Seo
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and Technology (KIST School)Korea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Miji Kwon
- Department of Smart Health Science and TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| | - June Hong Ahn
- Division of Pulmonology and AllergyDepartment of Internal MedicineCollege of MedicineYeungnam University and Regional Center for Respiratory DiseasesYeungnam University Medical CenterDaegu42415Republic of Korea
| | - Heungsoo Shin
- Department of BioengineeringHanyang UniversitySeoul04763Republic of Korea
| | - Gyu Yong Song
- College of PharmacyChungnam National UniversityDaejeon34134Republic of Korea
- AREZ Co. LtdDaejeon34134Republic of Korea
| | - Eun Ji Park
- D&D PharmatechSeongnam13486Republic of Korea
| | - Dong Hee Na
- College of PharmacyChung‐Ang UniversitySeoul06974Republic of Korea
| | - Seung‐Woo Cho
- Department of BiotechnologyYonsei UniversitySeoul03722Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and Technology (KIST School)Korea University of Science and Technology (UST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| | - Hee Ho Park
- Department of BioengineeringHanyang UniversitySeoul04763Republic of Korea
- BK21 FOUR Education and Research Group for Biopharmaceutical Innovation LeaderHanyang UniversitySeoul04763Republic of Korea
- Research Institute for Convergence of Basic ScienceHanyang UniversitySeoul04763Republic of Korea
| | - Wonhwa Lee
- Department of ChemistrySungkyunkwan UniversitySuwon16419Republic of Korea
| |
Collapse
|
4
|
Chen L, Huang Q, Zhao T, Sui L, Wang S, Xiao Z, Nan Y, Ai K. Nanotherapies for sepsis by regulating inflammatory signals and reactive oxygen and nitrogen species: New insight for treating COVID-19. Redox Biol 2021; 45:102046. [PMID: 34174559 PMCID: PMC8205260 DOI: 10.1016/j.redox.2021.102046] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 has caused up to 127 million cases of COVID-19. Approximately 5% of COVID-19 patients develop severe illness, and approximately 40% of those with severe illness eventually die, corresponding to more than 2.78 million people. The pathological characteristics of COVID-19 resemble typical sepsis, and severe COVID-19 has been identified as viral sepsis. Progress in sepsis research is important for improving the clinical care of these patients. Recent advances in understanding the pathogenesis of sepsis have led to the view that an uncontrolled inflammatory response and oxidative stress are core factors. However, in the traditional treatment of sepsis, it is difficult to achieve a balance between the inflammation, pathogens (viruses, bacteria, and fungi), and patient tolerance, resulting in high mortality of patients with sepsis. In recent years, nanomaterials mediating reactive oxygen and nitrogen species (RONS) and the inflammatory response have shown previously unattainable therapeutic effects on sepsis. Despite these advantages, RONS and inflammatory response-based nanomaterials have yet to be extensively adopted as sepsis therapy. To the best of our knowledge, no review has yet discussed the pathogenesis of sepsis and the application of nanomaterials. To help bridge this gap, we discuss the pathogenesis of sepsis related to inflammation and the overproduction RONS, which activate pathogen-associated molecular pattern (PAMP)-pattern recognition receptor (PRR) and damage-associated molecular pattern (DAMP)-PRR signaling pathways. We also summarize the application of nanomaterials in the treatment of sepsis. As highlighted here, this strategy could synergistically improve the therapeutic efficacy against both RONS and inflammation in sepsis and may prolong survival. Current challenges and future developments for sepsis treatment are also summarized.
Collapse
Affiliation(s)
- Li Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, Hunan, China
| | - Tianjiao Zhao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, Hunan, China
| | - Lihua Sui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Yayun Nan
- Geriatric Medical Center, Ningxia People's Hospital, Yinchuan, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.
| |
Collapse
|
5
|
Milanesi E, Manda G, Dobre M, Codrici E, Neagoe IV, Popescu BO, Bajenaru OA, Spiru L, Tudose C, Prada GI, Davidescu EI, Piñol-Ripoll G, Cuadrado A. Distinctive Under-Expression Profile of Inflammatory and Redox Genes in the Blood of Elderly Patients with Cardiovascular Disease. J Inflamm Res 2021; 14:429-442. [PMID: 33658823 PMCID: PMC7917358 DOI: 10.2147/jir.s280328] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/22/2020] [Indexed: 01/12/2023] Open
Abstract
Purpose Chronic low-grade inflammation and oxidative stress are present in most of the pathologic mechanisms underlying non-communicable diseases. Inflammation and redox biomarkers might therefore have a value in disease prognosis and therapy response. In this context, we performed a case–control study for assessing in whole blood the expression profile of inflammation and redox-related genes in elderly subjects with various comorbidities. Patients and Methods In the blood of 130 elderly subjects with various pathologies (cardiovascular disease, hypertension, dyslipidemia including hypercholesterolemia, type 2 diabetes mellitus), kept under control by polyvalent disease-specific medication, we investigated by pathway-focused qRT-PCR a panel comprising 84 inflammation-related and 84 redox-related genes. Results The study highlights a distinctive expression profile of genes critically involved in NF-κB-mediated inflammation and redox signaling in the blood of patients with cardiovascular disease, characterized by significant down-regulation of the genes NFKB2, NFKBIA, RELA, RELB, AKT1, IRF1, STAT1, CD40, LTA, TRAF2, PTGS1, ALOX12, DUOX1, DUOX2, MPO, GSR, TXNRD2, HSPA1A, MSRA, and PDLIM1. This gene expression profile defines the transcriptional status of blood leukocytes in stable disease under medication control, without discriminating between disease- and therapy-related changes. Conclusion The study brings preliminary proof on a minimally invasive strategy for monitoring disease in patients with cardiovascular pathology, from the point of view of inflammation or redox dysregulation in whole blood.
Collapse
Affiliation(s)
- Elena Milanesi
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Gina Manda
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Maria Dobre
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Elena Codrici
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | | | - Bogdan Ovidiu Popescu
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania.,Clinical Neurosciences, Geriatrics and Gerontology Departments, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 020021, Romania.,Neurology Department, Clinical Hospital Colentina, Bucharest, 020125, Romania
| | - Ovidiu Alexandru Bajenaru
- Clinical Neurosciences, Geriatrics and Gerontology Departments, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 020021, Romania.,Neurology Department, University Emergency Hospital, Bucharest, 050098, Romania
| | - Luiza Spiru
- Clinical Neurosciences, Geriatrics and Gerontology Departments, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 020021, Romania.,The Excellence Memory Center and Longevity Medicine, "Ana Aslan" International Foundation,, Bucharest, 050064, Romania
| | - Catalina Tudose
- Clinical Neurosciences, Geriatrics and Gerontology Departments, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 020021, Romania.,Section II, "Prof. Dr. Al. Obregia" Psychiatry Clinical Hospital & the Memory Center of the Romanian Alzheimer Society, Bucharest, 041914, Romania
| | - Gabriel-Ioan Prada
- Clinical Neurosciences, Geriatrics and Gerontology Departments, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 020021, Romania.,Section IV, "Ana Aslan" National Institute of Gerontology and Geriatrics, Bucharest, 011241, Romania
| | - Eugenia Irene Davidescu
- Clinical Neurosciences, Geriatrics and Gerontology Departments, "Carol Davila" University of Medicine and Pharmacy, Bucharest, 020021, Romania.,Neurology Department, Clinical Hospital Colentina, Bucharest, 020125, Romania
| | - Gerard Piñol-Ripoll
- Unitat Trastons Cognitius, Hospital Universitari Santa Maria-IRBLLeida, Lleida, 25198, Spain
| | - Antonio Cuadrado
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania.,Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain.,Neuroscience Section, Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, 28046, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain
| |
Collapse
|
6
|
Lasola JJM, Kamdem H, McDaniel MW, Pearson RM. Biomaterial-Driven Immunomodulation: Cell Biology-Based Strategies to Mitigate Severe Inflammation and Sepsis. Front Immunol 2020; 11:1726. [PMID: 32849612 PMCID: PMC7418829 DOI: 10.3389/fimmu.2020.01726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation is an essential component of a wide variety of disease processes and oftentimes can increase the deleterious effects of a disease. Finding ways to modulate this essential immune process is the basis for many therapeutics under development and is a burgeoning area of research for both basic and translational immunology. In addition to developing therapeutics for cellular and molecular targets, the use of biomaterials to modify innate and adaptive immune responses is an area that has recently sparked significant interest. In particular, immunomodulatory activity can be engineered into biomaterials to elicit heightened or dampened immune responses for use in vaccines, immune tolerance, or anti-inflammatory applications. Importantly, the inherent physicochemical properties of the biomaterials play a significant role in determining the observed effects. Properties including composition, molecular weight, size, surface charge, and others affect interactions with immune cells (i.e., nano-bio interactions) and allow for differential biological responses such as activation or inhibition of inflammatory signaling pathways, surface molecule expression, and antigen presentation to be encoded. Numerous opportunities to open new avenues of research to understand the ways in which immune cells interact with and integrate information from their environment may provide critical solutions needed to treat a variety of disorders and diseases where immune dysregulation is a key inciting event. However, to elicit predictable immune responses there is a great need for a thorough understanding of how the biomaterial properties can be tuned to harness a designed immunological outcome. This review aims to systematically describe the biological effects of nanoparticle properties-separate from additional small molecule or biologic delivery-on modulating innate immune cell responses in the context of severe inflammation and sepsis. We propose that nanoparticles represent a potential polypharmacological strategy to simultaneously modify multiple aspects of dysregulated immune responses where single target therapies have fallen short for these applications. This review intends to serve as a resource for immunology labs and other associated fields that would like to apply the growing field of rationally designed biomaterials into their work.
Collapse
Affiliation(s)
- Jackline Joy Martín Lasola
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Henry Kamdem
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Michael W. McDaniel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Ryan M. Pearson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
7
|
Einecke G, Beutel G, Hoeper MM, Kielstein JT. The answer is blowing in the wind: an uncommon cause for severe ARDS accompanied by circulatory insufficiency requiring extracorporeal membrane oxygenation. BMJ Case Rep 2017; 2017:bcr-2016-218079. [PMID: 28343152 DOI: 10.1136/bcr-2016-218079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We report a rare complication in an immunosuppressed patient with IgA nephropathy who suffered from severe acute respiratory distress syndrome, severe capillary leakage and shock after placement of a double lumen central venous catheter. He could be successfully treated by extracorporeal membrane oxygenation (ECMO) and therapeutic plasma exchange. This report highlights the severity of late-onset complications of catheter placements and shows the potential of ECMO treatment for the management of acute illnesses with bridge to recovery.
Collapse
Affiliation(s)
- Gunilla Einecke
- Department of Nephrology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Gernot Beutel
- Department of Hematology, Hemostaseology, Oncology and Stem Cell Transplantation, Medizinische Hochschule Hannover, Hannover, Germany
| | - Marius M Hoeper
- Department of Respiratory Medicine, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jan T Kielstein
- Department of Nephrology, Stadtisches Klinikum Braunschweig GmbH, Braunschweig, Germany
| |
Collapse
|
8
|
Lee W, Seo J, Kwak S, Park EJ, Na DH, Kim S, Lee YM, Kim IS, Bae JS. A Double-Chambered Protein Nanocage Loaded with Thrombin Receptor Agonist Peptide (TRAP) and γ-Carboxyglutamic Acid of Protein C (PC-Gla) for Sepsis Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:6637-6643. [PMID: 26414883 DOI: 10.1002/adma.201503093] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/20/2015] [Indexed: 06/05/2023]
Abstract
New protein nanocages are designed bearing two functional proteins, γ-carboxyglutamic acid of protein C (PC-Gla) and thrombin receptor agonist peptide (TRAP), and have an anti-septic response. These nanoparticles reduce sepsis-induced organ injury and septic mortality in vivo. Noting that there are currently no medications for severe sepsis, these results show that novel nanoparticles can be used to treat sepsis.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, South Korea
- Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, CMRI, School of Medicine, Kyungpook National University, Daegu, 700-422, South Korea
| | - Junyoung Seo
- Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, CMRI, School of Medicine, Kyungpook National University, Daegu, 700-422, South Korea
| | - Soyoung Kwak
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, South Korea
| | - Eun Ji Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, South Korea
| | - Dong Hee Na
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, South Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, CMRI, School of Medicine, Kyungpook National University, Daegu, 700-422, South Korea
| | - You Mie Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, South Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 136-791, South Korea
- KU-KIST School, Korea University, Seoul, 136-701, South Korea
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, South Korea
- Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, CMRI, School of Medicine, Kyungpook National University, Daegu, 700-422, South Korea
| |
Collapse
|
9
|
Kell DB, Pretorius E. The simultaneous occurrence of both hypercoagulability and hypofibrinolysis in blood and serum during systemic inflammation, and the roles of iron and fibrin(ogen). Integr Biol (Camb) 2015; 7:24-52. [PMID: 25335120 DOI: 10.1039/c4ib00173g] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although the two phenomena are usually studied separately, we summarise a considerable body of literature to the effect that a great many diseases involve (or are accompanied by) both an increased tendency for blood to clot (hypercoagulability) and the resistance of the clots so formed (hypofibrinolysis) to the typical, 'healthy' or physiological lysis. We concentrate here on the terminal stages of fibrin formation from fibrinogen, as catalysed by thrombin. Hypercoagulability goes hand in hand with inflammation, and is strongly influenced by the fibrinogen concentration (and vice versa); this can be mediated via interleukin-6. Poorly liganded iron is a significant feature of inflammatory diseases, and hypofibrinolysis may change as a result of changes in the structure and morphology of the clot, which may be mimicked in vitro, and may be caused in vivo, by the presence of unliganded iron interacting with fibrin(ogen) during clot formation. Many of these phenomena are probably caused by electrostatic changes in the iron-fibrinogen system, though hydroxyl radical (OH˙) formation can also contribute under both acute and (more especially) chronic conditions. Many substances are known to affect the nature of fibrin polymerised from fibrinogen, such that this might be seen as a kind of bellwether for human or plasma health. Overall, our analysis demonstrates the commonalities underpinning a variety of pathologies as seen in both hypercoagulability and hypofibrinolysis, and offers opportunities for both diagnostics and therapies.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess St, Manchester M1 7DN, Lancs, UK.
| | | |
Collapse
|
10
|
Luan ZG, Naranpurev M, Ma XC. Treatment of low molecular weight heparin inhibits systemic inflammation and prevents endotoxin-induced acute lung injury in rats. Inflammation 2015; 37:924-32. [PMID: 24425537 DOI: 10.1007/s10753-014-9812-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
To determine whether low molecular weight heparin (LMWH) is able to reduce pulmonary inflammation and improve the survival in rats with endotoxin-induced acute lung injury (ALI). Rat ALI model was reproduced by injection of lipopolysaccharide (LPS) into tail vein. Rats were divided randomly into three groups: control group, ALI group, LMWH-treated group. Blood was collected and lung tissue was harvested at the designated time points for analysis. The lung specimens were harvested for morphological studies, streptavidin-peroxidase immunohistochemistry examination. Lung tissue edema was evaluated by tissue water content. The levels of lung tissue myeloperoxidase (MPO) were determined. Meanwhile, the nuclear factor-kappa B (NF-κB) activation, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) levels and high mobility group box 1 (HMGB1) and intercellular adhesion molecule-1 (ICAM-1) protein levels in the lung were studied. In survival studies, a separate group of rats were treated with LMWH or sterile saline after LPS administration. Then, the mortality was recorded. Treatment with LMWH after ALI was associated with a reduction in the severity of LPS-induced lung injury. Treatment with LMWH significantly decreased the expression of TNF-α, IL-1β, HMGB1 and ICAM-1 in the lung of ALI rats. Similarly, treatment with LMWH dramatically diminished LPS-induced neutrophil sequestration and markedly reduced the enhanced lung permeability. In the present study, LMWH administration inhibited the nuclear translocation of NF-κB in the lung. Survival was significantly higher among the LMWH-treated group compared with the ALI group. These data suggest that LMWH attenuates inflammation and prevents lethality in endotoxemic rats.
Collapse
Affiliation(s)
- Zheng-Gang Luan
- Department of Intensive Care Unit, The First Hospital of China Medical University, 155 Nanjing North Street, Shenyang, 110001, Liaoning Province, China,
| | | | | |
Collapse
|
11
|
Madoiwa S. Recent advances in disseminated intravascular coagulation: endothelial cells and fibrinolysis in sepsis-induced DIC. J Intensive Care 2015; 3:8. [PMID: 27408725 PMCID: PMC4940964 DOI: 10.1186/s40560-015-0075-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 01/29/2015] [Indexed: 12/22/2022] Open
Abstract
Endothelial cells are highly active, sensing and responding to signals from extracellular environments. They act as gatekeepers, mediating the recruitment and extravasation of proinflammatory leukocytes to the sites of tissue damage or infection. Endothelial cells participate in fibrinolysis by secreting tissue-type plasminogen activator, which converts plasminogen to active enzyme plasmin through constitutive and regulated pathways. Fibrinolysis systems and inflammation are tightly linked, as both responses are major host defense systems against both healing processes of tissue repair as well as pathogenic microorganisms. Endothelial cell dysfunction is one of the early signs of systemic inflammation, and it is a trigger of multiple organ failure in sepsis. The marked increase in plasminogen activator inhibitor-1 level causes fibrinolytic shutdown in endotoxemia or sepsis and is one of the most important predictors of multiple organ dysfunction during sepsis-induced disseminated intravascular coagulation (DIC). Leukocytes exhibit the first-line response to microorganisms. Leukocyte-derived elastase degrades cross-linked fibrin to yield molecular species distinct from those generated by plasmin. The alternative systems for fibrinolysis that interact with the plasminogen activator-plasmin systems may play crucial roles in the lysis of fibrin clots in sepsis-induced DIC.
Collapse
Affiliation(s)
- Seiji Madoiwa
- Department of Clinical Laboratory Medicine, Tokyo Saiseikai Central Hospital, 1-14-17, Mita, Minato-ku, Tokyo 108-0073 Japan ; Department of Clinical Laboratory Medicine, Jichi Medical University, 3311-1, Yakushi-ji, Shimotsuke, Tochigi 329-0498 Japan
| |
Collapse
|
12
|
Rimmer E, Houston BL, Kumar A, Abou-Setta AM, Friesen C, Marshall JC, Rock G, Turgeon AF, Cook DJ, Houston DS, Zarychanski R. The efficacy and safety of plasma exchange in patients with sepsis and septic shock: a systematic review and meta-analysis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2014; 18:699. [PMID: 25527094 PMCID: PMC4318234 DOI: 10.1186/s13054-014-0699-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/27/2014] [Indexed: 01/04/2023]
Abstract
Introduction Sepsis and septic shock are leading causes of intensive care unit (ICU) mortality. They are characterized by excessive inflammation, upregulation of procoagulant proteins and depletion of natural anticoagulants. Plasma exchange has the potential to improve survival in sepsis by removing inflammatory cytokines and restoring deficient plasma proteins. The objective of this study is to evaluate the efficacy and safety of plasma exchange in patients with sepsis. Methods We searched MEDLINE, EMBASE, CENTRAL, Scopus, reference lists of relevant articles, and grey literature for relevant citations. We included randomized controlled trials comparing plasma exchange or plasma filtration with usual care in critically ill patients with sepsis or septic shock. Two reviewers independently identified trials, extracted trial-level data and performed risk of bias assessments using the Cochrane Risk of Bias tool. The primary outcome was all-cause mortality reported at longest follow-up. Meta-analysis was performed using a random-effects model. Results Of 1,957 records identified, we included four unique trials enrolling a total of 194 patients (one enrolling adults only, two enrolling children only, one enrolling adults and children). The mean age of adult patients ranged from 38 to 53 years (n = 128) and the mean age of children ranged from 0.9 to 18 years (n = 66). All trials were at unclear to high risk of bias. The use of plasma exchange was not associated with a significant reduction in all-cause mortality (risk ratio (RR) 0.83, 95% confidence interval (CI) 0.45 to 1.52, I2 60%). In adults, plasma exchange was associated with reduced mortality (RR 0.63, 95% CI 0.42 to 0.96; I2 0%), but was not in children (RR 0.96, 95% CI 0.28 to 3.38; I2 60%). None of the trials reported ICU or hospital lengths of stay. Only one trial reported adverse events associated with plasma exchange including six episodes of hypotension and one allergic reaction to fresh frozen plasma. Conclusions Insufficient evidence exists to recommend plasma exchange as an adjunctive therapy for patients with sepsis or septic shock. Rigorous randomized controlled trials evaluating clinically relevant patient-centered outcomes are required to evaluate the impact of plasma exchange in this condition. Electronic supplementary material The online version of this article (doi:10.1186/s13054-014-0699-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emily Rimmer
- Department of Internal Medicine, University of Manitoba, GC425-820 Sherbrook Street, HSC, Winnipeg, R3A 1R9, Canada. .,Department of Haematology and Medical Oncology, CancerCare Manitoba, 675 McDermot Ave, Winnipeg, R3E 0V9, Canada.
| | - Brett L Houston
- Faculty of Medicine, University of Manitoba, 250 Brodie Centre, 727 McDermot Ave, Winnipeg, R3E 3P5, Canada.
| | - Anand Kumar
- Department of Internal Medicine, University of Manitoba, GC425-820 Sherbrook Street, HSC, Winnipeg, R3A 1R9, Canada.
| | - Ahmed M Abou-Setta
- George & Fay Yee Center for Healthcare Innovation, University of Manitoba/Winnipeg Regional Health Authority, GE706-820 Sherbrook Street, HSC, Winnipeg, R3A 1R9, Canada.
| | - Carol Friesen
- Neil John Maclean Health Sciences Library, University of Manitoba, Brodie Centre, 727 McDermot Ave, Winnipeg, R3E 3P5, Canada.
| | - John C Marshall
- Section of Critical Care Medicine, St. Michael's Hospital, 30 Bond Street, Toronto, M5B 1W8, Canada.
| | - Gail Rock
- Ottawa Hospital, 501 Smyth Road, Ottawa, K1H 8L6, Canada.
| | - Alexis F Turgeon
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine and Population Health and Optimal Health Practices Unit, CHU de Québec Research Center, Université Laval, 1401-18th Street, Québec, G1J 1Z4, Canada.
| | - Deborah J Cook
- Department of Medicine, McMaster University, Hamilton, L8S 4K1, Canada. .,Department of Clinical Epidemiology and Biostatistics, McMaster University, 1280 Main Street, West, Hamilton, Canada.
| | - Donald S Houston
- Department of Internal Medicine, University of Manitoba, GC425-820 Sherbrook Street, HSC, Winnipeg, R3A 1R9, Canada. .,Department of Haematology and Medical Oncology, CancerCare Manitoba, 675 McDermot Ave, Winnipeg, R3E 0V9, Canada.
| | - Ryan Zarychanski
- Department of Internal Medicine, University of Manitoba, GC425-820 Sherbrook Street, HSC, Winnipeg, R3A 1R9, Canada. .,Department of Haematology and Medical Oncology, CancerCare Manitoba, 675 McDermot Ave, Winnipeg, R3E 0V9, Canada. .,George & Fay Yee Center for Healthcare Innovation, University of Manitoba/Winnipeg Regional Health Authority, GE706-820 Sherbrook Street, HSC, Winnipeg, R3A 1R9, Canada.
| |
Collapse
|
13
|
Shukla P, Rao GM, Pandey G, Sharma S, Mittapelly N, Shegokar R, Mishra PR. Therapeutic interventions in sepsis: current and anticipated pharmacological agents. Br J Pharmacol 2014; 171:5011-31. [PMID: 24977655 PMCID: PMC4253453 DOI: 10.1111/bph.12829] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/29/2014] [Accepted: 06/13/2014] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a clinical syndrome characterized by a multisystem response to a pathogenic assault due to underlying infection that involves a combination of interconnected biochemical, cellular and organ-organ interactive networks. After the withdrawal of recombinant human-activated protein C (rAPC), researchers and physicians have continued to search for new therapeutic approaches and targets against sepsis, effective in both hypo- and hyperinflammatory states. Currently, statins are being evaluated as a viable option in clinical trials. Many agents that have shown favourable results in experimental sepsis are not clinically effective or have not been clinically evaluated. Apart from developing new therapeutic molecules, there is great scope for for developing a variety of drug delivery strategies, such as nanoparticulate carriers and phospholipid-based systems. These nanoparticulate carriers neutralize intracorporeal LPS as well as deliver therapeutic agents to targeted tissues and subcellular locations. Here, we review and critically discuss the present status and new experimental and clinical approaches for therapeutic intervention in sepsis.
Collapse
Affiliation(s)
- Prashant Shukla
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - G Madhava Rao
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Gitu Pandey
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Shweta Sharma
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Naresh Mittapelly
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Ranjita Shegokar
- Department of Pharmaceutics, Biopharmaceutics & NutriCosmetics, Institute of Pharmacy, Freie Universität BerlinBerlin, Germany
| | - Prabhat Ranjan Mishra
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| |
Collapse
|
14
|
Li X, Liu Y, Wang L, Li Z, Ma X. Unfractionated heparin attenuates LPS-induced IL-8 secretion via PI3K/Akt/NF-κB signaling pathway in human endothelial cells. Immunobiology 2014; 220:399-405. [PMID: 25454806 DOI: 10.1016/j.imbio.2014.10.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 09/24/2014] [Accepted: 10/12/2014] [Indexed: 12/11/2022]
Abstract
Unfractionated heparin (UFH) is largely used as anti-thrombotic drug. While UFH has been shown to suppress lipopolysaccharide (LPS)-induced nuclear factor-κB (NF-κB) activation, intracellular upstream events that cause NF-κB down-regulation in response to UFH remain unclear. Thus, we investigated the involvement of phosphoinositide-3-OH kinase (PI3K)/Akt in the inhibition of LPS-activated NF-κB pathway by UFH in human pulmonary microvascular endothelial cells (HPMECs). Pretreatment with UFH (0.1-1U/ml) significantly inhibited LPS (10μg/ml)-stimulated interleukin (IL)-6 and IL-8 production in HPMECs. LPS activated Akt and NF-κB, whereas UFH suppresses LPS-induced Akt phosphorylation and NF-κB nuclear translocation, which were required for IL-6 and IL-8 gene transcription. Inhibition studies by using wortmannin abrogated NF-κB-mediated IL-6 and IL-8 expression, suggesting the requirement of PI3K/Akt pathway. Our data provided the first evidence that UFH might repress LPS-activated PI3K/Akt pathway, leading to inhibitory effect of NF-κB activation with diminished IL-6 and IL-8 expression in HPMECs.
Collapse
Affiliation(s)
- Xu Li
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China.
| | - Yina Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Liang Wang
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Zhiliang Li
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Xiaochun Ma
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China.
| |
Collapse
|
15
|
Li X, Li X, Zheng Z, Liu Y, Ma X. Unfractionated heparin suppresses lipopolysaccharide-induced monocyte chemoattractant protein-1 expression in human microvascular endothelial cells by blocking Krüppel-like factor 5 and nuclear factor-κB pathway. Immunobiology 2014; 219:778-85. [PMID: 25023776 DOI: 10.1016/j.imbio.2014.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/24/2014] [Indexed: 11/17/2022]
Abstract
Unfractionated heparin (UFH) and low-molecular-weight heparins (LMWH), apart from anticoagulant activities, contain a variety of biological properties such as anti-inflammatory actions possibly affecting sepsis. Chemokines are vital for promoting the movement of circulating leukocytes to the site of infection and are involved in the pathogenesis of sepsis. The purpose of this study was to investigate the effects and potential mechanisms of UFH on lipopolysaccharide (LPS)-induced chemokine production in human pulmonary microvascular endothelial cells (HPMECs). HPMECs were pretreated with UFH (0.1 U/ml and 1 U/ml), 15 min prior to stimulation with LPS (10 μg/ml). Cells were cultured under various experimental conditions for 2 h and 6 h for analysis. UFH markedly decreased LPS-induced interleukin (IL)-8 and monocyte chemoattractant protein-1 (MCP-1) mRNA and protein expression in HPMECs. UFH also attenuated the secretion of these chemokines in culture supernatants. In addition, UFH blocked the chemotactic activities of LPS-stimulated HPMECs supernatants on monocytes migration as expected. UFH inhibited LPS-induced Krüppel-like factor 5 (KLF-5) mRNA and protein levels. Concurrently, UFH reduced nuclear factor (NF)-κB nuclear translocation. Importantly, transfection with siRNA targeting KLF-5 reduced NF-κB activation and chemokines expression. These results demonstrate that interfering with KLF-5 mediated NF-κB activation might contribute to the inhibitory effects of chemokines and monocytes migration by UFH in LPS-stimulated HPMECs.
Collapse
Affiliation(s)
- Xu Li
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Xin Li
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Zhen Zheng
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Yina Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China
| | - Xiaochun Ma
- Department of Intensive Care Unit, The First Affiliated Hospital, China Medical University, Bei-er Road 92, Shenyang 110001, Liaoning Province, PR China.
| |
Collapse
|
16
|
Abstract
During the past 15 years, several anti-inflammatory treatments have failed to reduce mortality in patients with severe sepsis. However, recent evidence indicates that coagulation abnormalities in sepsis may play a major role in the pathogenesis of multiple organ failure and the high mortality rate in patients with severe sepsis. Interestingly, blockade of the coagulant pathway can inhibit both procoagulant and proinflammatory pathways in sepsis. Protein C, a natural anticoagulant, interrupts several of the pathophysiologic pathways in sepsis. Acquired protein C deficiency is present in the majority of septic patients and is associated with unfavorable outcomes. Protein C replacement therapy was effective in preclinical animal models of sepsis in reducing end-organ damage and mortality. Recent clinical trials of protein C replacement in human meningococcemia resulted in a markedly decreased morbidity and mortality. And, most importantly, in a recently completed large, randomized trial of activated protein C treatment in severe sepsis, mortality was reduced from 30.8% in the placebo group to 24.7% in the treatment group at 28 days. Thus, there is new evidence that mortality can be reduced among patients with severe sepsis through the use of a new therapy that inhibits the procoagulant and the inflammatory cascades.
Collapse
Affiliation(s)
- Mark R Looney
- Cardiovascular Research Institute, University of California, 505 Parnassus Avenue, 94143-0130, San Francisco, CA, USA
| | | |
Collapse
|
17
|
Deutschman CS. Therapy for sepsis: Einstein once said... Br J Anaesth 2013; 111:693-5. [PMID: 24108726 DOI: 10.1093/bja/aet157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- C S Deutschman
- Perelman School of Medicine at the University of Pennsylvania - Anesthesiology and Critical Care, Dulles 781A/HUP, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| |
Collapse
|
18
|
MicroRNA expression following activated protein C treatment during septic shock. J Surg Res 2013; 182:116-26. [DOI: 10.1016/j.jss.2012.07.063] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 07/06/2012] [Accepted: 07/25/2012] [Indexed: 12/12/2022]
|
19
|
Machado RJA, Monteiro NKV, Migliolo L, Silva ON, Pinto MFS, Oliveira AS, Franco OL, Kiyota S, Bemquerer MP, Uchoa AF, Morais AHA, Santos EA. Characterization and pharmacological properties of a novel multifunctional Kunitz inhibitor from Erythrina velutina seeds. PLoS One 2013; 8:e63571. [PMID: 23737945 PMCID: PMC3666885 DOI: 10.1371/journal.pone.0063571] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/04/2013] [Indexed: 11/19/2022] Open
Abstract
Inhibitors of peptidases isolated from leguminous seeds have been studied for their pharmacological properties. The present study focused on purification, biochemical characterization and anti-inflammatory and anticoagulant evaluation of a novel Kunitz trypsin inhibitor from Erythrina velutina seeds (EvTI). Trypsin inhibitors were purified by ammonium sulfate (30-60%), fractionation followed by Trypsin-Sepharose affinity chromatography and reversed-phase high performance liquid chromatography. The purified inhibitor showed molecular mass of 19,210.48 Da. Furthermore, a second isoform with 19,228.16 Da was also observed. The inhibitor that showed highest trypsin specificity and enhanced recovery yield was named EvTI (P2) and was selected for further analysis. The EvTI peptide fragments, generated by trypsin and pepsin digestion, were further analyzed by MALDI-ToF-ToF mass spectrometry, allowing a partial primary structure elucidation. EvTI exhibited inhibitory activity against trypsin with IC50 of 2.2×10(-8) mol.L(-1) and constant inhibition (Ki) of 1.0×10(-8) mol.L(-1), by a non-competitive mechanism. In addition to inhibit the activity of trypsin, EvTI also inhibited factor Xa and neutrophil elastase, but do not inhibit thrombin, chymotrypsin or peptidase 3. EvTI was investigated for its anti-inflammatory and anti-coagulant properties. Firstly, EvTI showed no cytotoxic effect on human peripheral blood cells. Nevertheless, the inhibitor was able to prolong the clotting time in a dose-dependent manner by using in vitro and in vivo models. Due to anti-inflammatory and anticoagulant EvTI properties, two sepsis models were here challenged. EvTI inhibited leukocyte migration and specifically acted by inhibiting TNF-α release and stimulating IFN-α and IL-12 synthesis. The data presented clearly contribute to a better understanding of the use of Kunitz inhibitors in sepsis as a bioactive agent capable of interfering in blood coagulation and inflammation.
Collapse
Affiliation(s)
- Richele J. A. Machado
- Laboratório de Química e Função de Proteínas Bioativas, Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brasil
| | - Norberto K. V. Monteiro
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, RN, Brasil
| | - Ludovico Migliolo
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Gênomicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasil
| | - Osmar N. Silva
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Gênomicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasil
| | - Michele F. S. Pinto
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Gênomicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasil
| | - Adeliana S. Oliveira
- Laboratório de Química e Função de Proteínas Bioativas, Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brasil
| | - Octávio L. Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Gênomicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brasil
| | - Sumika Kiyota
- Laboratório de Bioquímica de Proteínas e Peptídeos, Centro de Pesquisa e Desenvolvimento de Sanidade Animal, Instituto Biológico, São Paulo, Brasil
| | - Marcelo P. Bemquerer
- Laboratório de Espectrometria de Massa, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Brasil
| | - Adriana F. Uchoa
- Departamento de Biologia Celular e Genética, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brasil
| | - Ana H. A. Morais
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, RN, Brasil
| | - Elizeu A. Santos
- Laboratório de Química e Função de Proteínas Bioativas, Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brasil
| |
Collapse
|
20
|
Abstract
BACKGROUND Septic shock is a highly inflammatory and procoagulant state associated with significant mortality. In a single randomized controlled trial, recombinant human activated protein C (drotrecogin alfa) reduced mortality in patients with severe sepsis at high risk of death. Further clinical trials, including a recently completed trial in patients with septic shock, failed to reproduce these results. OBJECTIVE To evaluate the effectiveness of recombinant human activated protein C on mortality in a cohort of patients with septic shock and to explore possible reasons for inconsistent results in previous studies. DESIGN Retrospective, 2:1 propensity-matched, multicenter cohort study. SETTING Twenty-nine academic and community intensive care units in three countries. PATIENTS Seven thousand three hundred ninety-two adult patients diagnosed with septic shock, of which 349 received recombinant human activated protein C within 48 hrs of intensive care unit admission between 1997 and 2007. MEASUREMENTS AND MAIN RESULTS Our primary outcomes were mortality over 30 days and mortality stratified by Acute Physiology and Chronic Health Evaluation II quartile. Using a propensity-matched Cox proportional hazard model, we observed a 6.1% absolute reduction in 30-day mortality associated with the use of recombinant human activated protein C (108/311 [34.7%] vs. 254/622 [40.8%], hazard ratio 0.72, 95% confidence interval 0.52-1.00, p = .05) and noted consistent reductions in mortality among Acute Physiology and Chronic Health Evaluation II quartiles. A time to event analysis showed that the time to appropriate antimicrobials after documented hypotension decreased for each year of study (p = .003), a finding that was congruent with a decrease in annual mortality over the study period (odds ratio 0.96 per year [95% confidence interval 0.93-0.99], p = .003). CONCLUSIONS In this retrospective, propensity-matched, multicenter cohort study of patients with septic shock, early use of recombinant human activated protein C was associated with reduced mortality. Improvements in general quality of care such as speed of antimicrobial delivery leading to decreasing mortality of patients with septic shock may have contributed to the null results of the recently completed trial of recombinant human activated protein C in patients with septic shock.
Collapse
|
21
|
Unfractionated heparin inhibits lipopolysaccharide-induced inflammatory response through blocking p38 MAPK and NF-κB activation on endothelial cell. Cytokine 2012; 60:114-21. [DOI: 10.1016/j.cyto.2012.06.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 05/29/2012] [Accepted: 06/04/2012] [Indexed: 11/17/2022]
|
22
|
Heeb MJ, Marzec U, Gruber A, Hanson SR. Antithrombotic activity of protein S infused without activated protein C in a baboon thrombosis model. Thromb Haemost 2012; 107:690-8. [PMID: 22370911 DOI: 10.1160/th11-10-0699] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/12/2012] [Indexed: 12/15/2022]
Abstract
Protein S (ProS) is an essential plasma protein that enhances the anticoagulant activity of activated protein C (APC). In vitro , purified native human Zn2+-containing ProS also exerts direct anticoagulant activity by inhibiting prothrombinase and extrinsic FXase activities independently of APC. We investigated antithrombotic effects of ProS infused without APC in a baboon shunt model of thrombogenesis that employs a device consisting of arterial and venous shear flow segments. In in vitro experiments, the Zn2+-containing human ProS used for the studies displayed >10-fold higher prothrombinase inhibitory activity and anticoagulant activity in tissue factor-stimulated plasma, and four-fold higher inhibition of the intrinsic pathway than the Zn2+-deficient ProS used. In the thrombosis model, ProS (33 μg/minute for 1 hour) or saline was infused locally; platelet and fibrin deposition in the shunt were measured over 2 hours. During experiments performed at 50 ml/minute blood flow, Zn2+-containing ProS inhibited platelet deposition 73-96% in arterial-type flow segments and 90-99% in venous-type flow segments; Zn2+-deficient ProS inhibited platelet deposition 52% in arterial-type flow segments and 65-73% in venous-type flow segments. At 100 ml/min blood flow rate, Zn2+-containing ProS inhibited platelet deposition by 39% and 73% in the respective segments; Zn2+-deficient ProS inhibited platelet deposition by 5% and 0% in the respective segments. Zn2+-containing ProS suppressed fibrin deposition by 67-90%. Systemic APC-independent ProS activity was significantly increased and thrombin-antithrombin complex levels were significantly decreased after infusion of ProS. Thus, infused human Zn2+-containing ProS is antithrombotic in primates, and may have therapeutic potential even in protein C-deficient human patients.
Collapse
Affiliation(s)
- M J Heeb
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
23
|
Avery JW, Smith GM, Owino SO, Sarr D, Nagy T, Mwalimu S, Matthias J, Kelly LF, Poovassery JS, Middii JD, Abramowsky C, Moore JM. Maternal malaria induces a procoagulant and antifibrinolytic state that is embryotoxic but responsive to anticoagulant therapy. PLoS One 2012; 7:e31090. [PMID: 22347435 PMCID: PMC3274552 DOI: 10.1371/journal.pone.0031090] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 01/02/2012] [Indexed: 01/06/2023] Open
Abstract
Low birth weight and fetal loss are commonly attributed to malaria in endemic areas, but the cellular and molecular mechanisms that underlie these poor birth outcomes are incompletely understood. Increasing evidence suggests that dysregulated hemostasis is important in malaria pathogenesis, but its role in placental malaria (PM), characterized by intervillous sequestration of Plasmodium falciparum, proinflammatory responses, and excessive fibrin deposition is not known. To address this question, markers of coagulation and fibrinolysis were assessed in placentae from malaria-exposed primigravid women. PM was associated with significantly elevated placental monocyte and proinflammatory marker levels, enhanced perivillous fibrin deposition, and increased markers of activated coagulation and suppressed fibrinolysis in placental plasma. Submicroscopic PM was not proinflammatory but tended to be procoagulant and antifibrinolytic. Birth weight trended downward in association with placental parasitemia and high fibrin score. To directly assess the importance of coagulation in malaria-induced compromise of pregnancy, Plasmodium chabaudi AS-infected pregnant C57BL/6 mice were treated with the anticoagulant, low molecular weight heparin. Treatment rescued pregnancy at midgestation, with substantially decreased rates of active abortion and reduced placental and embryonic hemorrhage and necrosis relative to untreated animals. Together, the results suggest that dysregulated hemostasis may represent a novel therapeutic target in malaria-compromised pregnancies.
Collapse
Affiliation(s)
- John W. Avery
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Geoffrey M. Smith
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Simon O. Owino
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Demba Sarr
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Tamas Nagy
- Department of Veterinary Pathology, University of Georgia, Athens, Georgia, United States of America
| | - Stephen Mwalimu
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - James Matthias
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia, United States of America
| | - Lauren F. Kelly
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Jayakumar S. Poovassery
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Joab D. Middii
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Carlos Abramowsky
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Julie M. Moore
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
24
|
Enoxaparin and fondaparinux attenuates endothelial damage in endotoxemic rats. J Trauma Acute Care Surg 2012; 72:177-82. [DOI: 10.1097/ta.0b013e31821a83f0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
25
|
Le urgenze ematologiche. ITALIAN JOURNAL OF MEDICINE 2011. [DOI: 10.1016/j.itjm.2010.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
26
|
Abstract
OBJECTIVES The aim of this present study was to investigate the d-dimer in acute pancreatitis and its associations with triglyceride (TG). METHODS The d-dimer was measured in 45 patients with mild acute pancreatitis, 43 patients with severe acute pancreatitis, and 45 healthy controls. Eighty-eight patients were divided into high and low TG groups based on their TG levels. Twenty outpatients with serumal TG levels higher than 5.65 mM were chosen as hypertriglyceridemia controls. We investigated whether there were any correlations between the d-dimer levels and serumal TG in acute pancreatitis. RESULTS In 45 patients with mild acute pancreatitis, the d-dimer increased to approximately 2 times over the reference value, whereas in 43 patients with severe acute pancreatitis, the d-dimer level increased to 6 times above the limit; the difference was significant. Both TG and acute pancreatitis could cause an elevation of the d-dimer level, in which TG takes a more important role. The increase in the d-dimer was also directly related to the severity of acute pancreatitis. CONCLUSIONS Plasma concentrations of the d-dimer increase in acute pancreatitis. The increase in TG is probably the main cause of the d-dimer elevation in patients with acute pancreatitis.
Collapse
|
27
|
Protective effect and mechanism of sodium tanshinone II A sulfonate on microcirculatory disturbance of small intestine in rats with sepsis. ACTA ACUST UNITED AC 2011; 31:441. [PMID: 21823002 DOI: 10.1007/s11596-011-0470-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Indexed: 10/17/2022]
Abstract
To explore the protective effect of sodium tanshinone IIA sulfonate (STS) on microcirculatory disturbance of small intestine in rats with sepsis, and the possible mechanism, a rat model of sepsis was induced by cecal ligation and puncture (CLP). Rats were randomly divided into 3 groups: sham operated group (S), sepsis group (CLP) and STS treatment group (STS). STS (1 mg/kg) was slowly injected through the right external jugular vein after CLP. The histopathologic changes in the intestinal tissue and changes of mesenteric microcirculation were observed. The levels of tumor necrosis factor-α (TNF-α) in the intestinal tissue were determined by using enzyme-linked immunoabsorbent assay (ELISA). The expression of intercellular adhesion molecule-1 (ICAM-1) in the intestinal tissue was detected by using immunohistochemisty and Western blot, that of nuclear factor κB (NF-κB) and tissue factor (TF) by using Western blot, and the levels of NF-κB mRNA expression by using RT-PCR respectively. The microcirculatory disturbance of the intestine was aggravated after CLP. The injury of the intestinal tissues was obviously aggravated in CLP group as compared with S group. The expression levels of NF-κB p65, ICAM-1, TF and TNF-α were upregulaed after CLP (P<0.01). STS post-treatment could ameliorate the microcirculatory disturbance, attenuate the injury of the intestinal tissues induced by CLP, and decrease the levels of NF-κB, ICAM-1, TF and TNF-α (P<0.01). It is suggested that STS can ameliorate the microcirculatory disturbance of the small intestine in rats with sepsis, and the mechanism may be associated with the inhibition of inflammatory responses and amelioration of coagulation abnormality.
Collapse
|
28
|
Zhang WJ, Wei H, Tien YT, Frei B. Genetic ablation of phagocytic NADPH oxidase in mice limits TNFα-induced inflammation in the lungs but not other tissues. Free Radic Biol Med 2011; 50:1517-25. [PMID: 21376114 PMCID: PMC3090478 DOI: 10.1016/j.freeradbiomed.2011.02.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 02/18/2011] [Accepted: 02/24/2011] [Indexed: 11/19/2022]
Abstract
In vitro and limited in vivo evidence suggests that reactive oxygen species derived from NADPH oxidases (NOX-ROS) play an important role in inflammatory responses by enhancing the activity of redox-sensitive cell signaling pathways and transcription factors. Here, we investigated the role of NOX-ROS in TNFα-induced acute inflammatory responses in vivo, using mice deficient in the gp91(phox) (NOX2) or p47(phox) subunits of NADPH oxidase. Age- and body weight-matched C57BL/6J wild-type (WT) and gp91(phox) or p47(phox) knockout mice were injected intraperitoneally with 50 μg TNFα/kg bw or saline vehicle control and sacrificed at various time points up to 24 h. Compared to WT mice, gp91(phox -/-) mice exhibited significantly diminished (P<0.05) TNFα-induced acute inflammatory responses in the lungs but not other tissues, including heart, liver, and kidney, as evidenced by decreased activation of the redox-sensitive transcription factor NF-κB, and decreased gene expression of interleukin (IL)-1β, IL-6, TNFα, E-selectin, and other cellular adhesion molecules. Similar results were observed in p47(phox -/-) mice. Interestingly, decreased lung inflammation in knockout mice was accompanied by increased leukocyte infiltration into the lungs compared to other tissues. Our data suggest that phagocytic NOX-ROS signaling plays a critical role in promoting TNFα-induced, NF-κB-dependent acute inflammatory responses and tissue injury specifically in the lungs, which is effected by preferential leukocyte infiltration.
Collapse
Affiliation(s)
- Wei-Jian Zhang
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- Correspondence and reprint requests should be addressed to: Wei-Jian Zhang and Balz Frei, Linus Pauling Institute, Oregon State University, 571 Weniger Hall, Corvallis, OR 97331, Phone: (541) 737-5075, FAX: (541) 737-5077, and
| | - Hao Wei
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Ying-Tzang Tien
- Histopathology Laboratory, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Balz Frei
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- Correspondence and reprint requests should be addressed to: Wei-Jian Zhang and Balz Frei, Linus Pauling Institute, Oregon State University, 571 Weniger Hall, Corvallis, OR 97331, Phone: (541) 737-5075, FAX: (541) 737-5077, and
| |
Collapse
|
29
|
Ahmed TAN, Karalis I, Jukema JW. Emerging drugs for coronary artery disease. From past achievements and current needs to clinical promises. Expert Opin Emerg Drugs 2011; 16:203-33. [DOI: 10.1517/14728214.2011.549606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Yada N, Nishio K, Seki T, Fukushima H, Urizono Y, Hata M, Okuchi K. Therapeutic effect of recombinant thrombomodulin on the hemostatic and inflammatory response in patients with septic DIC. ACTA ACUST UNITED AC 2011. [DOI: 10.3893/jjaam.22.749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
AAV/hSTAT3-gene delivery lowers aortic inflammatory cell infiltration in LDLR KO mice on high cholesterol. Atherosclerosis 2010; 213:59-66. [DOI: 10.1016/j.atherosclerosis.2010.07.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 11/23/2022]
|
32
|
|
33
|
IN VIVO AND IN VITRO EFFECTS OF THE ANTICOAGULANT, THROMBOMODULIN, ON THE INFLAMMATORY RESPONSE IN RODENT MODELS. Shock 2010; 33:282-8. [DOI: 10.1097/shk.0b013e3181b0ef7b] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
34
|
Horstman LL, Jy W, Ahn YS, Zivadinov R, Maghzi AH, Etemadifar M, Steven Alexander J, Minagar A. Role of platelets in neuroinflammation: a wide-angle perspective. J Neuroinflammation 2010; 7:10. [PMID: 20128908 PMCID: PMC2829540 DOI: 10.1186/1742-2094-7-10] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 02/03/2010] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES This review summarizes recent developments in platelet biology relevant to neuroinflammatory disorders. Multiple sclerosis (MS) is taken as the "Poster Child" of these disorders but the implications are wide. The role of platelets in inflammation is well appreciated in the cardiovascular and cancer research communities but appears to be relatively neglected in neurological research. ORGANIZATION After a brief introduction to platelets, topics covered include the matrix metalloproteinases, platelet chemokines, cytokines and growth factors, the recent finding of platelet PPAR receptors and Toll-like receptors, complement, bioactive lipids, and other agents/functions likely to be relevant in neuroinflammatory diseases. Each section cites literature linking the topic to areas of active research in MS or other disorders, including especially Alzheimer's disease. CONCLUSION The final section summarizes evidence of platelet involvement in MS. The general conclusion is that platelets may be key players in MS and related disorders, and warrant more attention in neurological research.
Collapse
Affiliation(s)
- Lawrence L Horstman
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Wenche Jy
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Yeon S Ahn
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, The Jacobs Neurological Institute, Department of Neurology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo NY, USA
| | - Amir H Maghzi
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - J Steven Alexander
- Department of Cellular and Molecular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
35
|
Akay S, Ozutemiz O, Yenisey C, Genc Simsek N, Yuce G, Batur Y. Use of activated protein C has no avail in the early phase of acute pancreatitis. HPB (Oxford) 2008; 10:459-63. [PMID: 19088933 PMCID: PMC2597320 DOI: 10.1080/13651820802140729] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Sepsis and acute pancreatitis have similar pathogenetic mechanisms that have been implicated in the progression of multiple organ failure. Drotrecogin alfa, an analogue of endogenous protein C, reduces mortality in clinical sepsis. Our objective was to evaluate the early therapeutic effects of activated protein C (APC) in a rat model of acute necrotizing pancreatitis. SUBJECTS AND METHOD Acute necrotizing pancreatitis was induced by intraductal injection of 5% Na taurocholate. Hourly bolus injections of saline or recombinant human APC (drotrecogin alfa) was commenced via femoral venous catheter four hours after the induction of acute pancreatitis. The experiment was terminated nine hours after pancreatitis induction. Animals in group one (n=20) had a sham operation while animals in group two (n=20) received saline and animals in group three (n=20) received drotrecogin alfa boluses after acute pancreatitis induction. Pancreatic tissue for histopathologic scores and myeloperoxidase, glutathione reductase, glutathione peroxidase, and catalase activities were collected, and blood for serum amylase, urea, creatinine, and interleukin-6 measurements was withdrawn. RESULTS Serum amylase activity was significantly lower in the APC treated group than the untreated group (17,435+/-432 U/L vs. 27,426+/-118 U/L, respectively). While the serum interleukin-6 concentration in the APC untreated group was significantly lower than the treated group (970+/-323 pg/mL vs. 330+/-368 pg/mL, respectively). CONCLUSION In the early phase of acute pancreatitis, drotrecogin alfa treatment did not result in a significant improvement in oxidative and inflammatory parameters or renal functions.
Collapse
Affiliation(s)
- Sinan Akay
- Ege University Hospital Gastroenterology DepartmentIzmirTurkey
| | - Omer Ozutemiz
- Ege University Hospital Gastroenterology DepartmentIzmirTurkey
| | - Cigdem Yenisey
- Adnan Menderes University Biochemistry DepartmentAydinTurkey
| | | | - Gul Yuce
- Ege University Hospital Pathology DepartmentIzmirTurkey
| | - Yucel Batur
- Ege University Hospital Gastroenterology DepartmentIzmirTurkey
| |
Collapse
|
36
|
Finfer S, Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Gårdlund B, Marshall JC, Rhodes A. Design, conduct, analysis and reporting of a multi-national placebo-controlled trial of activated protein C for persistent septic shock. Intensive Care Med 2008; 34:1935-47. [PMID: 18839141 PMCID: PMC2995439 DOI: 10.1007/s00134-008-1266-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 08/19/2008] [Indexed: 12/22/2022]
Abstract
The role of drotrecogin alfa (activated) (DAA) in severe sepsis remains controversial and clinicians are unsure whether or not to treat their patients with DAA. In response to a request from the European Medicines Agency, Eli Lilly will sponsor a new placebo-controlled trial and history suggests the results will be subject to great scrutiny. An academic steering committee will oversee the conduct of the study and will write the study manuscripts. The steering committee intends that the study will be conducted with the maximum possible transparency; this includes publication of the study protocol and a memorandum of understanding which delineates the role of the sponsor. The trial has the potential to provide clinicians with valuable data but patients will only benefit if clinicians have confidence in the conduct, analysis and reporting of the trial. This special article describes the process by which the trial was developed, major decisions regarding trial design, and plans for independent analysis, interpretation and reporting of the data.
Collapse
Affiliation(s)
- Simon Finfer
- Critical Care and Trauma, The George Institute for International Health, University of Sydney, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
ASHP Guidelines on Medication Cost Management Strategies for Hospitals and Health Systems. Am J Health Syst Pharm 2008; 65:1368-84. [DOI: 10.2146/ajhp080021] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
38
|
Kalil AC, Sun J. How many patients with severe sepsis are needed to confirm the efficacy of drotrecogin alfa activated? A Bayesian design. Intensive Care Med 2008; 34:1804-11. [PMID: 18504550 DOI: 10.1007/s00134-008-1159-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 05/08/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Controversies concerning the pivotal trial and negative results from other Drotrecogin alfa activated (DAA) trials have raised questions about its efficacy in severe sepsis patients. Based on all available evidence, our study aimed to respond to: (1) What is the current probability that DAA is not better than the control? If the current probability is not small, e.g. greater than 0.05, then (2) How many patients will be needed for the DAA confirmatory trial? DESIGN We applied the Bayesian methodology to all randomized trials which tested anti-thrombotic therapies for severe sepsis. Prior distributions were defined as severe skeptic (all anti-thrombotic trials or DAA trials); moderate skeptic (anti-thrombotic trials with high risk of death); mild skeptic (DAA trials only); enthusiastic (DAA trials with high risk of death). RESULTS The total study sample includes 7,683 patients: DAA (N = 3,143); Anti-thrombin-III (N = 2,581); tissue factor pathway inhibitor (N = 1,959). Answer 1: All current probabilities that DAA is not better than the control (except for the enthusiastic analysis) range from 0.14 to 0.48, which strongly suggest the need for a confirmatory trial. Answer 2: The number of patients necessary for the DAA confirmatory trial ranges up to 8,350 for the severe skeptic; 730-810 for moderate skeptic; 550-685 for mild skeptic; and zero for enthusiastic analysis. CONCLUSIONS A confirmatory trial with approximately 600 patients with severe sepsis and high risk of death can provide a convincing answer for both the mild and moderate skeptic physicians concerning the efficacy of DAA in severe sepsis.
Collapse
Affiliation(s)
- Andre C Kalil
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| | | |
Collapse
|
39
|
Hagiwara S, Iwasaka H, Hidaka S, Hishiyama S, Noguchi T. Danaparoid sodium inhibits systemic inflammation and prevents endotoxin-induced acute lung injury in rats. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 12:R43. [PMID: 18380908 PMCID: PMC2447588 DOI: 10.1186/cc6851] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 02/05/2008] [Accepted: 04/02/2008] [Indexed: 02/06/2023]
Abstract
Introduction Systemic inflammatory mediators, including high mobility group box 1 (HMGB1), play an important role in the development of sepsis. Anticoagulants, such as danaparoid sodium (DA), may be able to inhibit sepsis-induced inflammation, but the mechanism of action is not well understood. We hypothesised that DA would act as an inhibitor of systemic inflammation and prevent endotoxin-induced acute lung injury in a rat model. Methods We used male Wistar rats. Animals in the intervention arm received a bolus of 50 U/kg of DA or saline injected into the tail vein after lipopolysaccharide (LPS) administration. We measured cytokine (tumour necrosis factor (TNF)α, interleukin (IL)-6 and IL-10) and HMGB1 levels in serum and lung tissue at regular intervals for 12 h following LPS injection. The mouse macrophage cell line RAW 264.7 was assessed following stimulation with LPS alone or concurrently with DA with identification of HMGB1 and other cytokines in the supernatant. Results Survival was significantly higher and lung histopathology significantly improved among the DA (50 U/kg) animals compared to the control rats. The serum and lung HMGB1 levels were lower over time among DA-treated animals. In the in vitro study, administration of DA was associated with decreased production of HMGB1. In the cell signalling studies, DA administration inhibited the phosphorylation of IκB. Conclusion DA decreases cytokine and HMGB1 levels during LPS-induced inflammation. As a result, DA ameliorated lung pathology and reduces mortality in endotoxin-induced systemic inflammation in a rat model. This effect may be mediated through the inhibition of cytokines and HMGB1.
Collapse
Affiliation(s)
- Satoshi Hagiwara
- Department of Brain and Nerve Science, Anesthesiology, Oita University Faculty of Medicine, Oita, Japan.
| | | | | | | | | |
Collapse
|
40
|
Liu F, Liu Y, Lui VC, Lamb JR, Tam PK, Chen Y. Hypoxia modulates lipopolysaccharide induced TNF-α expression in murine macrophages. Exp Cell Res 2008; 314:1327-36. [DOI: 10.1016/j.yexcr.2008.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 01/08/2008] [Accepted: 01/08/2008] [Indexed: 01/22/2023]
|
41
|
Predicting the severity of systemic inflammatory response syndrome (SIRS)-associated coagulopathy with hemostatic molecular markers and vascular endothelial injury markers. ACTA ACUST UNITED AC 2008; 63:1093-8. [PMID: 17993956 DOI: 10.1097/01.ta.0000251420.41427.d3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The changes in biomarkers of coagulation or fibrinolysis, anticoagulation, inflammation, and endothelial damage occur in patients with systemic inflammatory response syndrome (SIRS). The purpose of this study is to assess the prognostic value of these markers in patients with SIRS-associated hypercoagulopathy. METHODS Sixty-six SIRS patients with a platelet count less than 15.0 x 10(4)/mm3 in three university hospital intensive care units were enrolled in this prospective, comparative study. Blood samples were obtained on day 0 and day 2. Twelve hemostatic, inflammatory, and vascular endothelial indices were measured and the data were compared between the severe group (patients with a total maximum Sequential Organ Failure Assessment score of 10 or more and nonsurvivors; n = 25) and the less-severe group (Sequential Organ Failure Assessment score <10; n = 41). RESULTS Significant changes between the groups were observed in platelet count, fibrin or fibrinogen degradation products, interleukin-6, soluble thrombomodulin, antithrombin (AT) activity, and protein C activity, both on day 0 and on day 2. In contrast, the d-dimer, soluble fibrin, plasmin-[alpha]2-antiplasmin complex, and E-selectin levels were higher in the severe group only on day 2. No significant difference was seen regarding the thrombin-AT complex and total plasminogen activator inhibitor on both days. A comparison of the areas under the receiver operating characteristic curve revealed the AT activity to be the best predictor of a progression of organ dysfunction. CONCLUSION The changes in some hemostatic molecular markers and vascular endothelial markers were conspicuous in patients with organ dysfunction. The AT activity is considered to be the most useful predictor of organ dysfunction.
Collapse
|
42
|
High dose antithrombin III inhibits HMGB1 and improves endotoxin-induced acute lung injury in rats. Intensive Care Med 2007; 34:361-7. [PMID: 17940748 DOI: 10.1007/s00134-007-0887-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Accepted: 09/17/2007] [Indexed: 12/18/2022]
Abstract
OBJECTIVE High mobility group box 1 (HMGB1) is an important factor in the development of sepsis. Previous work suggests that antithrombin III (ATIII) inhibits inflammation, but the mechanism of action is still poorly understood. DESIGN AND SETTING Prospective controlled animal study in a university laboratory. MATERIALS Rats were randomly divided into a lipopolysaccharide (LPS)-induced sepsis control group and an ATIII-treated experimental group. Animals in the experimental group received a bolus of 250 units/kg of ATIII injected into the tail vein. MEASUREMENTS AND RESULTS Animals receiving high-dose ATIII (250 units/kg) had significantly improved lung histopathology and survival compared to the control rats. We measured serum and lung levels of various cytokines and HMGB1 at regular intervals from 0 to 12 h after the induction of sepsis and demonstrated lower HMGB1 levels over time in ATIII-treated animals. In an in vitro experiment, we stimulated the mouse macrophage cell line RAW 264.7 with LPS in the presence or absence of ATIII. Subsequent measurement of HMGB1 concentrations in the supernatant and cell signaling molecules in cell lysates revealed an ATIII dose-dependent decrease in HMGB1 release. Furthermore, inhibition of IkB and p42 phosphorylation was observed with the administration of ATIII, suggestive of downstream signaling pathways. CONCLUSIONS High-dose ATIII decreases lung pathology and reduces mortality in a rat sepsis model. This finding may be mediated by the inhibition of HMGB1.
Collapse
|
43
|
Al-Sabah S, Goldberg P, Qureshi ST. Pasteurella multocida septic shock following liver transplantation treated with drotrecogin alfa (activated). Transpl Infect Dis 2007; 9:233-6. [PMID: 17692071 DOI: 10.1111/j.1399-3062.2006.00192.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Severe sepsis and progression to septic shock in solid organ transplant recipients is associated with a high mortality. We describe a fulminant case of septic shock in a liver transplant recipient caused by Pasteurella multocida, a gram-negative coccobacillus most commonly associated with domestic cats and dogs. P. multocida is a rare cause of bacteremia and has not been reported as a cause of septic shock following liver transplantation. In addition to standard therapy, the patient was managed with drotrecogin alpha (activated) recombinant activated protein C (APC), an evidence-based agent that has been shown to significantly improve outcome in severe sepsis in the non-transplant population. The known risk factors, clinical course, and outcomes of severe infection associated with P. multocida are also briefly reviewed. This case illustrates the need for transplant recipients and their healthcare providers to carefully consider the risk of severe infection associated with domestic animal exposure.
Collapse
Affiliation(s)
- S Al-Sabah
- Department of Surgery, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
44
|
Rice TW. Treatment of severe sepsis: where next? Current and future treatment approaches after the introduction of drotrecogin alfa. Vasc Health Risk Manag 2007; 2:3-18. [PMID: 17319465 PMCID: PMC1993976 DOI: 10.2147/vhrm.2006.2.1.3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Severely septic patients continue to experience excessive morbidity and mortality despite recent advances in critical care. Although significant resources have been invested in new treatments, almost all have failed to improve outcomes. An improved understanding of sepsis pathophysiology, including the complex interactions between inflammatory, coagulation, and fibrinolytic systems, has accelerated the development of novel treatments. Recombinant human activated protein C (rhAPC), or drotrecogin alfa (activated) (DAA), is currently the only US Food and Drug Administration (FDA)-approved medicine for the treatment of severe sepsis, and only in patients with a high risk of death. This review will discuss the treatment of severe sepsis, focusing on recent discoveries and unresolved questions about DAA's optimal use. Increasing pharmacological experience has generated enthusiasm for investigating medicines already approved for other indications as treatments for severe sepsis. Replacement doses of hydrocortisone and vasopressin may reduce mortality and improve hypotension, respectively, in a subgroup of patients with catecholamine-refractory septic shock. In addition to discussing these new indications, this review will detail the provocative preliminary data from four promising treatments, including two novel modalities: antagonizing high mobility group box protein and inhibiting tissue factor (TF). Observational data from the uncontrolled administration of heparin or statins in septic patients will also be reviewed.
Collapse
Affiliation(s)
- Todd W Rice
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville,TN 37232-2650, USA.
| |
Collapse
|
45
|
Watanabe R, Matsumoto T, Sano G, Ishii Y, Tateda K, Sumiyama Y, Uchiyama J, Sakurai S, Matsuzaki S, Imai S, Yamaguchi K. Efficacy of bacteriophage therapy against gut-derived sepsis caused by Pseudomonas aeruginosa in mice. Antimicrob Agents Chemother 2007; 51:446-52. [PMID: 17116686 PMCID: PMC1797723 DOI: 10.1128/aac.00635-06] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Revised: 06/23/2006] [Accepted: 11/07/2006] [Indexed: 12/30/2022] Open
Abstract
We evaluated the efficacy of bacteriophage (phage) therapy by using a murine model of gut-derived sepsis caused by Pseudomonas aeruginosa that closely resembles the clinical pathophysiology of septicemia in humans. Oral administration of a newly isolated lytic phage strain (KPP10) significantly protected mice against mortality (survival rates, 66.7% for the phage-treated group versus 0% for the saline-treated control group; P<0.01). Mice treated with phage also had lower numbers of viable P. aeruginosa cells in their blood, liver, and spleen. The levels of inflammatory cytokines (tumor necrosis factor alpha TNF-alpha, interleukin-1beta [IL-1beta], and IL-6) in blood and liver were significantly lower in phage-treated mice than in phage-untreated mice. The number of viable P. aeruginosa cells in fecal matter in the gastrointestinal tract was significantly lower in phage-treated mice than in the saline-treated control mice. We also studied the efficacy of phage treatment for intraperitoneal infection caused by P. aeruginosa and found that phage treatment significantly improved the survival of mice, but only under limited experimental conditions. In conclusion, our findings suggest that oral administration of phage may be effective against gut-derived sepsis caused by P. aeruginosa.
Collapse
Affiliation(s)
- Ryohei Watanabe
- Third Department of Surgery, Toho University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Severe sepsis and septic shock are common causes of morbidity and mortality. Interventions directed at specific endpoints, when initiated early in the "golden hours" of patient arrival at the hospital, seem to be promising. Early hemodynamic optimization, administration of appropriate antimicrobial therapy, and effective source control of infection are the cornerstones of successful management. In patients with vasopressor-dependent septic shock, provision of physiologic doses of replacement steroids may result in improved survival. Administration of drotrecogin alfa (activated), (activated protein C) has been shown to improve survival in patients with severe sepsis and septic shock who have a high risk of mortality. In this article we review the multi-modality approach to early diagnosis and intervention in the therapy of patients with severe sepsis and septic shock.
Collapse
Affiliation(s)
- Murugan Raghavan
- Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
47
|
Werdan K. Disseminated intravascular coagulation: A matter for immune globulins in sepsis?*. Crit Care Med 2006; 34:2505-6. [PMID: 16810102 DOI: 10.1097/01.ccm.0000230383.45428.c3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Triantafilou K, Triantafilou M. Innate recognition of bacteria: potential therapeutic targets for bacterial sepsis. Expert Rev Clin Immunol 2006; 2:787-99. [PMID: 20477633 DOI: 10.1586/1744666x.2.5.787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Sepsis is a paradoxical and complex disorder that results from an over-reaction of our innate immune system to bacterial infections. Although this disorder has been known since ancient times, the history of clinical research into novel therapies for sepsis has been disappointing. The inability to translate our findings to the clinic could be attributed to our lack of knowledge of the molecular mechanisms involved in sensing microbial pathogens. However, in the last decade, the innate immune sensors responsible for triggering this disease have been discovered. This review will examine mediators that have been targeted in the past, as well as in the present, and propose novel therapeutic interventions for the future.
Collapse
Affiliation(s)
- Kathy Triantafilou
- Infection and Immunity Group, University of Sussex, School of Life Sciences, Falmer, Brighton, BN1 9QG, UK.
| | | |
Collapse
|
49
|
Martin SR, Foley MR. Intensive care in obstetrics: an evidence-based review. Am J Obstet Gynecol 2006; 195:673-89. [PMID: 16949397 DOI: 10.1016/j.ajog.2006.05.042] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 03/05/2006] [Accepted: 05/30/2006] [Indexed: 11/26/2022]
|
50
|
Affiliation(s)
- Zacharyx A Rubin
- Division of Infectious Diseases, Emory University School of Medicine and Grady Memorial Hospital, Atlanta, Georgia 30303, USA
| | | | | |
Collapse
|