1
|
Brunialti MKC, Leite GGF, Eburneo GS, de Araujo OR, Peçanha-Pietrobom PM, Ferreira PRA, Bellei NCJ, Arakaki JSO, Medina-Pestana J, Requião-Moura L, Salomao R. Patterns of Circulating Cytokines and Vascular Markers' Response in the Presence of COVID-19 in Kidney Transplant Recipients Compared with Non-Transplanted Patients. Viruses 2023; 15:2166. [PMID: 38005844 PMCID: PMC10675241 DOI: 10.3390/v15112166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
COVID-19's severity has been associated with a possible imbalance in the cross-regulation of cytokines and vascular mediators. Since the beginning of the pandemic, kidney transplant recipients (KTRs) have been identified as patients of high vulnerability to more severe diseases. Thus, aiming to describe the patterns of cytokines and vascular mediators and to trace patients' differences according to their KTR status, this prospective study enrolled 67 COVID-19 patients (20 KTRs) and 29 non-COVID-19 controls before vaccination. A panel comprising 17 circulating cytokines and vascular mediators was run on samples collected at different time points. The cytokine and mediator patterns were investigated via principal component analysis (PCA) and correlation-based network (CBN). In both groups, compared to their respective controls, COVID-19 was associated with higher levels of cytokines and vascular mediators. Differentiating between the KTRs and non-KTRs, the number of correlations was much higher in the non-KTRs (44 vs. 14), and the node analysis showed the highest interactions of NGAL and sVCAM-1 in the non-KTRs and KTRs (9 vs. 4), respectively. In the PCA, while the non-KTRs with COVID-19 were differentiated from their controls in their IL-10, IFN-α, and TNF-α, this pattern was marked in the NGAL, sVCAM-1, and IL-8 of the KTRs.
Collapse
Affiliation(s)
- Milena Karina Coló Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Giuseppe G. F. Leite
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Gabriela Strafolino Eburneo
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Orlei Ribeiro de Araujo
- Intensive Care Unit, GRAACC, Pediatric Institute of Oncology, Universidade Federal de São Paulo, São Paulo 04039-001, Brazil;
| | - Paula M. Peçanha-Pietrobom
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Paulo Roberto Abrão Ferreira
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Nancy C. Junqueira Bellei
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Jaquelina Sonoe Ota Arakaki
- Division of Respiratory Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04020-050, Brazil;
| | - José Medina-Pestana
- Division of Nephrology, Universidade Federal de São Paulo, São Paulo 04038-031, Brazil;
- Hospital do Rim, Fundação Oswalado Ramos, São Paulo 04038-002, Brazil
| | - Lúcio Requião-Moura
- Division of Nephrology, Universidade Federal de São Paulo, São Paulo 04038-031, Brazil;
- Hospital do Rim, Fundação Oswalado Ramos, São Paulo 04038-002, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
- Hospital São Paulo, São Paulo 04024-002, Brazil
| |
Collapse
|
2
|
Brennan KA, Bhutiani M, Kingeter MA, McEvoy MD. Updates in the Management of Perioperative Vasoplegic Syndrome. Adv Anesth 2022; 40:71-92. [PMID: 36333053 DOI: 10.1016/j.aan.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Vasoplegic syndrome occurs relatively frequently in cardiac surgery, liver transplant, major noncardiac surgery, in post-return of spontaneous circulation situations, and in pateints with sepsis. It is paramount for the anesthesiologist to understand both the pathophysiology of vasoplegia and the different treatment strategies available for rescuing a patient from life-threatening hypotension.
Collapse
Affiliation(s)
- Kaitlyn A Brennan
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 21st Avenue South, MAB 422, Nashville, TN 37212, USA
| | - Monica Bhutiani
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 21st Avenue South, VUH 4107, Nashville, TN 37212, USA
| | - Meredith A Kingeter
- Anesthesia Residency, Vanderbilt University Medical Center, 1215 21st Avenue South, Suite 5160 MCE NT, Nashville, TN 37212, USA
| | - Matthew D McEvoy
- VUMC Enhanced Recovery Programs, Department of Anesthesiology, Vanderbilt University Medical Center, 1301 Medical Center Drive, TVC 4648, Nashville, TN 37232, USA.
| |
Collapse
|
3
|
Vitiello A, La Porta R, Ferrara F. The Role of Vitamin C in the Treatment of Sepsis. DRUGS & THERAPY PERSPECTIVES 2022. [DOI: 10.1007/s40267-022-00895-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
4
|
Wang Z, Huang J, Xie D, He D, Lu A, Liang C. Toward Overcoming Treatment Failure in Rheumatoid Arthritis. Front Immunol 2021; 12:755844. [PMID: 35003068 PMCID: PMC8732378 DOI: 10.3389/fimmu.2021.755844] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by inflammation and bone erosion. The exact mechanism of RA is still unknown, but various immune cytokines, signaling pathways and effector cells are involved. Disease-modifying antirheumatic drugs (DMARDs) are commonly used in RA treatment and classified into different categories. Nevertheless, RA treatment is based on a "trial-and-error" approach, and a substantial proportion of patients show failed therapy for each DMARD. Over the past decades, great efforts have been made to overcome treatment failure, including identification of biomarkers, exploration of the reasons for loss of efficacy, development of sequential or combinational DMARDs strategies and approval of new DMARDs. Here, we summarize these efforts, which would provide valuable insights for accurate RA clinical medication. While gratifying, researchers realize that these efforts are still far from enough to recommend specific DMARDs for individual patients. Precision medicine is an emerging medical model that proposes a highly individualized and tailored approach for disease management. In this review, we also discuss the potential of precision medicine for overcoming RA treatment failure, with the introduction of various cutting-edge technologies and big data.
Collapse
Affiliation(s)
- Zhuqian Wang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Duoli Xie
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Dongyi He
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Chao Liang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Nassiri AA, Hakemi MS, Miri MM, Shahrami R, Koomleh AA, Sabaghian T. Blood purification with CytoSorb in critically ill COVID-19 patients: A case series of 26 patients. Artif Organs 2021; 45:1338-1347. [PMID: 34152629 PMCID: PMC8444787 DOI: 10.1111/aor.14024] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/30/2022]
Abstract
Severe forms of the coronavirus disease 2019 (COVID‐19) can progress to sepsis‐like complications accompanied by “cytokine storm” for which the most effective treatment has not yet been established. Our study describes the results of CytoSorb hemoadsorption in COVID‐19 patients treated on the intensive care unit (ICU). In this retrospective study, 26 patients with COVID‐19 and acute respiratory distress syndrome (ARDS) were treated with hemoadsorption therapy. Pre‐, and post‐treatment values (clinical and laboratory) were compared. Data are expressed as mean (confidence intervals, CI), or median [interquartile ranges, IQR], as appropriate. Patients received 2 hemoadsorption treatments. This resulted in a significant decrease in norepinephrine requirements, and inflammatory marker plasma concentrations (procalcitonin, C‐reactive protein, ferritin) when comparing pre versus post treatment levels. The PaO2/FiO2 and overall organ function (ie, Sequential Organ Failure Assessment—SOFA score) also improved significantly. Patients stayed on the ICU for 9 days and 21 of them survived. To the best of our knowledge, this is one of the largest case series to date reporting early experiences on extracorporeal hemoadsorption therapy in SARS‐CoV‐2 positive patients with hyperinflammation and moderate ARDS. Treatment proved to be effective, technically feasible and well‐tolerated.
Collapse
Affiliation(s)
- Amir Ahmad Nassiri
- Division of Nephrology, Department of Internal Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Monir Sadat Hakemi
- Nephrology ward, Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mir Mohammad Miri
- Anesthesiology and Critical Care Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Shahrami
- Intensive Care Department, NIOC Tehran Hospital, Tehran, Iran
| | - Azadeh Ahmadi Koomleh
- Division of Nephrology, Department of Internal Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Sabaghian
- Division of Nephrology, Department of Internal Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Bhavani SV, Wolfe KS, Hrusch CL, Greenberg JA, Krishack PA, Lin J, Lecompte-Osorio P, Carey KA, Kress JP, Coopersmith CM, Sperling AI, Verhoef PA, Churpek MM, Patel BK. Temperature Trajectory Subphenotypes Correlate With Immune Responses in Patients With Sepsis. Crit Care Med 2021; 48:1645-1653. [PMID: 32947475 DOI: 10.1097/ccm.0000000000004610] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES We recently found that distinct body temperature trajectories of infected patients correlated with survival. Understanding the relationship between the temperature trajectories and the host immune response to infection could allow us to immunophenotype patients at the bedside using temperature. The objective was to identify whether temperature trajectories have consistent associations with specific cytokine responses in two distinct cohorts of infected patients. DESIGN Prospective observational study. SETTING Large academic medical center between 2013 and 2019. SUBJECTS Two cohorts of infected patients: 1) patients in the ICU with septic shock and 2) hospitalized patients with Staphylococcus aureus bacteremia. INTERVENTIONS Clinical data (including body temperature) and plasma cytokine concentrations were measured. Patients were classified into four temperature trajectory subphenotypes using their temperature measurements in the first 72 hours from the onset of infection. Log-transformed cytokine levels were standardized to the mean and compared with the subphenotypes in both cohorts. MEASUREMENTS AND MAIN RESULTS The cohorts consisted of 120 patients with septic shock (cohort 1) and 88 patients with S. aureus bacteremia (cohort 2). Patients from both cohorts were classified into one of four previously validated temperature subphenotypes: "hyperthermic, slow resolvers" (n = 19 cohort 1; n = 13 cohort 2), "hyperthermic, fast resolvers" (n = 18 C1; n = 24 C2), "normothermic" (n = 54 C1; n = 31 C2), and "hypothermic" (n = 29 C1; n = 20 C2). Both "hyperthermic, slow resolvers" and "hyperthermic, fast resolvers" had high levels of G-CSF, CCL2, and interleukin-10 compared with the "hypothermic" group when controlling for cohort and timing of cytokine measurement (p < 0.05). In contrast to the "hyperthermic, slow resolvers," the "hyperthermic, fast resolvers" showed significant decreases in the levels of several cytokines over a 24-hour period, including interleukin-1RA, interleukin-6, interleukin-8, G-CSF, and M-CSF (p < 0.001). CONCLUSIONS Temperature trajectory subphenotypes are associated with consistent cytokine profiles in two distinct cohorts of infected patients. These subphenotypes could play a role in the bedside identification of cytokine profiles in patients with sepsis.
Collapse
Affiliation(s)
| | - Krysta S Wolfe
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| | - Cara L Hrusch
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| | | | | | - Julie Lin
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| | | | - Kyle A Carey
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| | - John P Kress
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| | | | - Anne I Sperling
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| | | | - Matthew M Churpek
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
| | - Bhakti K Patel
- Department of Medicine, University of Chicago Medical Center, Chicago, IL
| |
Collapse
|
7
|
Keshari RS, Silasi R, Popescu NI, Regmi G, Chaaban H, Lambris JD, Lupu C, Mollnes TE, Lupu F. CD14 inhibition improves survival and attenuates thrombo-inflammation and cardiopulmonary dysfunction in a baboon model of Escherichia coli sepsis. J Thromb Haemost 2021; 19:429-443. [PMID: 33174372 PMCID: PMC8312235 DOI: 10.1111/jth.15162] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/02/2020] [Accepted: 10/26/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND During sepsis, gram-negative bacteria induce robust inflammation primarily via lipopolysacharride (LPS) signaling through TLR4, a process that involves the glycosylphosphatidylinositol (GPI)-anchored receptor CD14 transferring LPS to the Toll-like receptor 4/myeloid differentiation factor 2 (TLR4/MD-2) complex. Sepsis also triggers the onset of disseminated intravascular coagulation and consumptive coagulopathy. OBJECTIVES We investigated the effect of CD14 blockade on sepsis-induced coagulopathy, inflammation, organ dysfunction, and mortality. METHODS We used a baboon model of lethal Escherichia (E) coli sepsis to study two experimental groups (n = 5): (a) E coli challenge; (b) E coli challenge plus anti-CD14 (23G4) inhibitory antibody administered as an intravenous bolus 30 minutes before the E coli. RESULTS Following anti-CD14 treatment, two animals reached the 7-day end-point survivor criteria, while three animals had a significantly prolonged survival as compared to the non-treated animals that developed multiple organ failure and died within 30 hours. Anti-CD14 reduced the activation of coagulation through inhibition of tissue factor-dependent pathway, especially in the survivors, and enhanced the fibrinolysis due to strong inhibition of plasminogen activator inhibitor 1. The treatment prevented the robust complement activation induced by E coli, as shown by significantly decreased C3b, C5a, and sC5b-9. Vital signs, organ function biomarkers, bacteria clearance, and leukocyte and fibrinogen consumption were all improved at varying levels. Anti-CD14 reduced neutrophil activation, cell death, LPS levels, and pro-inflammatory cytokines (tumor necrosis factor, interleukin (IL)-6, IL-1β, IL-8, interferon gamma, monocyte chemoattractant protein-1), more significantly in the survivors than non-surviving animals. CONCLUSIONS Our results highlight the crosstalk between coagulation/fibrinolysis, inflammation, and complement systems and suggest a protective role of anti-CD14 treatment in E coli sepsis.
Collapse
Affiliation(s)
- Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Narcis I. Popescu
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Girija Regmi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hala Chaaban
- Department of Pediatrics, Neonatal and Perinatal Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - John D. Lambris
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tom E. Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway
- Research Laboratory Nordland Hospital, K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Bodo, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Departments of Cell Biology, Pathology and Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
8
|
Monneret G, Benlyamani I, Gossez M, Bermejo-Martin JF, Martín-Fernandez M, Sesques P, Wallet F, Venet F. COVID-19: What type of cytokine storm are we dealing with? J Med Virol 2020; 93:197-198. [PMID: 32681651 PMCID: PMC7405026 DOI: 10.1002/jmv.26317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 01/24/2023]
Affiliation(s)
- Guillaume Monneret
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression (PI3)," Claude Bernard Lyon 1 University Hospices Civils de Lyon-bioMérieux, Lyon, France.,Immunology Department, Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Ihsane Benlyamani
- Immunology Department, Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Morgane Gossez
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression (PI3)," Claude Bernard Lyon 1 University Hospices Civils de Lyon-bioMérieux, Lyon, France.,Immunology Department, Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Jesus F Bermejo-Martin
- Biomedical research Department, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain.,Biomedical research Department, Hospital Universitario Río Hortega de Valladolid, Valladolid, Spain
| | - Marta Martín-Fernandez
- Biomedical research Department, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain.,Biomedical research Department, Hospital Universitario Río Hortega de Valladolid, Valladolid, Spain
| | - Pierre Sesques
- Medical Intensive Care Unit, Hospices Civils de Lyon, Lyon Sud Teaching Hospital, Lyon, France
| | - Florent Wallet
- Department of Hematology, Hospices Civils de Lyon, Lyon Sud Teaching Hospital, Lyon, France
| | - Fabienne Venet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression (PI3)," Claude Bernard Lyon 1 University Hospices Civils de Lyon-bioMérieux, Lyon, France.,Immunology Department, Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
9
|
Meng F, Du C, Zhang Y, Wang S, Zhou Q, Wu L, Wang Y, Yang X. Protective effect of rhubarb combined with ulinastatin for patients with sepsis. Medicine (Baltimore) 2020; 99:e18895. [PMID: 32049789 PMCID: PMC7035124 DOI: 10.1097/md.0000000000018895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Sepsis is the leading cause of death in critically ill patients. Ulinastatin (UTI), a protease inhibitor, and rhubarb, used as a traditional Chinese medication, are proved to be effective in treating sepsis, but the effect of the combination therapy of these two drugs on sepsis remains unclear. This study aimed to investigate the effect of the combination treatment of UTI and rhubarb on sepsis patients. METHODS A total of 75 septic patients were randomly divided into control group, UTI group, Rhubarb group, and UTI plus Rhubarb group. Clinical data and score of Acute Physiology and Chronic Health Evaluation II (APACHE II) were collected; lymphocyte subtypes in the peripheral blood were analyzed before and after the 5-day treatment in the Intensive Care Unit. RESULTS All the therapeutic interventions (UTI alone, rhubarb alone, or UTI plus rhubarb) significantly reduced the levels of C-Reactive protein, white blood cell density, lactic acid, and APACH II scores, and elevated the levels of CD4/CD8, but only UTI plus rhubarb treatment obviously decreased the level of procalcitonin. CONCLUSION This study suggested that the combination of UTI and rhubarb may be a promising therapeutic scheme to ameliorate sepsis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanli Wang
- Department of Mental Health, First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | | |
Collapse
|
10
|
Abstract
OBJECTIVES Sterile and infectious critical illnesses often result in vasoplegic shock and a robust systemic inflammatory response that are similar in presentation. The innate immune system is at the center of the response to both infectious and traumatic insults. Damage-associated molecular patterns are small molecules that are released from stressed or dying cells. Damage-associated molecular patterns activate pattern recognition receptors and coordinate the leading edge of the innate immune response. This review introduces the concept of damage-associated molecular patterns and how they activate a systemic inflammatory response, specifically in trauma, neurologic injury, and infection. It also explores how, when carried to extremes, damage-associated molecular patterns may even perpetuate multisystem organ failure. DATA SOURCES Basic and clinical studies were obtained from a PubMed search through August 2017. STUDY SELECTION Articles considered include original articles, review articles, and conference proceedings. DATA EXTRACTION An analysis of scientific, peer-reviewed data was performed. High quality preclinical and clinical studies adjudicated by the authors were included and summarized. DATA SYNTHESIS Pattern recognition receptors respond to damage-associated molecular patterns and then activate inflammatory pathways. Damage-associated molecular patterns have been linked to the recruitment of sentinel leukocytes and the initiation of the inflammatory cascade. Damage-associated molecular patterns have been linked to many conditions in critical care illnesses. Preclinical models have added insight into how they may mediate distant organ dysfunction. CONCLUSIONS Damage-associated molecular pattern activation and release is an important research for intensive care practitioners. It will add to our understanding of the phase and state of the innate immune response to an insult. Early work is encouraging. However, only with improved understanding of damage-associated molecular pattern activation and function, we can perhaps hope to target damage-associated molecular patterns as diagnostic and/or therapeutic modalities in the future.
Collapse
|
11
|
Spiller F, Oliveira Formiga R, Fernandes da Silva Coimbra J, Alves-Filho JC, Cunha TM, Cunha FQ. Targeting nitric oxide as a key modulator of sepsis, arthritis and pain. Nitric Oxide 2019; 89:32-40. [PMID: 31051258 DOI: 10.1016/j.niox.2019.04.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/22/2019] [Accepted: 04/29/2019] [Indexed: 12/25/2022]
Abstract
Nitric oxide (NO) is produced by enzymatic activity of neuronal (nNOS), endothelial (eNOS), and inducible nitric oxide synthase (iNOS) and modulates a broad spectrum of physiological and pathophysiological conditions. The iNOS isoform is positively regulated at transcriptional level and produces high levels of NO in response to inflammatory mediators and/or to pattern recognition receptor signaling, such as Toll-like receptors. In this review, we compiled the main contributions of our group for understanding of the role of NO in sepsis and arthritis outcome and the peripheral contributions of NO to inflammatory pain development. Although neutrophil iNOS-derived NO is necessary for bacterial killing, systemic production of high levels of NO impairs neutrophil migration to infections through inhibiting neutrophil adhesion on microcirculation and their locomotion. Moreover, neutrophil-derived NO contributes to multiple organ dysfunction in sepsis. In arthritis, NO is chief for bacterial clearance in staphylococcal-induced arthritis; however, it contributes to articular damage and bone mass degradation. NO produced in inflammatory sites also downmodulates pain. The mechanism involved in analgesic effect and inhibition of neutrophil migration is dependent on the activation of the classical sGC/cGMP/PKG pathway. Despite the increasing number of studies performed after the identification of NO as an endothelium-derived relaxing factor, the underlying mechanisms of NO in inflammatory diseases remain unclear.
Collapse
Affiliation(s)
- Fernando Spiller
- Department of Pharmacology, Federal University of Santa Catarina (UFSC), Florianopolis, Brazil.
| | | | | | | | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeiro Preto Medical School, University of Sao Paulo, Brazil
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeiro Preto Medical School, University of Sao Paulo, Brazil.
| |
Collapse
|
12
|
Salomão R, Ferreira B, Salomão M, Santos S, Azevedo L, Brunialti M. Sepsis: evolving concepts and challenges. Braz J Med Biol Res 2019; 52:e8595. [PMID: 30994733 PMCID: PMC6472937 DOI: 10.1590/1414-431x20198595] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
Sepsis remains a major cause of morbidity and mortality worldwide, with increased burden in low- and middle-resource settings. The role of the inflammatory response in the pathogenesis of the syndrome has supported the modern concept of sepsis. Nevertheless, a definition of sepsis and the criteria for its recognition is a continuous process, which reflects the growing knowledge of its mechanisms and the success and failure of diagnostic and therapeutic interventions. Here we review the evolving concepts of sepsis, from the "systemic inflammatory response syndrome triggered by infection" (Sepsis-1) to "a severe, potentially fatal, organic dysfunction caused by an inadequate or dysregulated host response to infection" (Sepsis-3). We focused in the pathophysiology behind the concept and the criteria for recognition and diagnosis of sepsis. A major challenge in evaluating the host response in sepsis is to characterize what is protective and what is harmful, and we discuss that, at least in part, the apparent dysregulated host response may be an effort to adapt to a hostile environment. The new criteria for recognition and diagnosis of sepsis were derived from robust databases, restricted, however, to developed countries. Since then, the criteria have been supported in different clinical settings and in different economic and epidemiological contexts, but still raise discussion regarding their use for the identification versus the prognostication of the septic patient. Clinicians should not be restricted to definition criteria when evaluating patients with infection and should wisely use the broad array of information obtained by rigorous clinical observation.
Collapse
Affiliation(s)
- R. Salomão
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - B.L. Ferreira
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M.C. Salomão
- Departamento de Moléstias Infecciosas e Parasitárias Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - S.S. Santos
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L.C.P. Azevedo
- Unidade de Terapia Intensiva do Hospital Sírio Libanês, São Paulo, SP, Brasil
| | - M.K.C. Brunialti
- Disciplina de Infectologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
13
|
Pimienta G, Heithoff DM, Rosa-Campos A, Tran M, Esko JD, Mahan MJ, Marth JD, Smith JW. Plasma Proteome Signature of Sepsis: a Functionally Connected Protein Network. Proteomics 2019; 19:e1800389. [PMID: 30706660 DOI: 10.1002/pmic.201800389] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/14/2019] [Indexed: 12/29/2022]
Abstract
Sepsis is an extreme host response to infection that leads to loss of organ function and cardiovascular integrity. Mortality from sepsis is on the rise. Despite more than three decades of research and clinical trials, specific diagnostic and therapeutic strategies for sepsis are still absent. The use of LFQ- and TMT-based quantitative proteomics is reported here to study the plasma proteome in five mouse models of sepsis. A knowledge-based interpretation of the data reveals a protein network with extensive connectivity through documented functional or physical interactions. The individual proteins in the network all have a documented role in sepsis and are known to be extracellular. The changes in protein abundance observed in the mouse models of sepsis have for the most part the same directionality (increased or decreased abundance) as reported in the literature for human sepsis. This network has been named the Plasma Proteome Signature of Sepsis (PPSS). The PPSS is a quantifiable molecular readout that can supplant the current symptom-based approach used to diagnose sepsis. This type of molecular interpretation of sepsis, its progression, and its response to therapeutic intervention are an important step in advancing our understanding of sepsis, and for discovering and evaluating new therapeutic strategies.
Collapse
Affiliation(s)
- Genaro Pimienta
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA
| | - Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.,Center for Nanomedicine, University of California, Santa Barbara, CA, 93106, USA
| | - Alexandre Rosa-Campos
- Proteomics Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Minerva Tran
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA
| | - Jamey D Marth
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.,Center for Nanomedicine, University of California, Santa Barbara, CA, 93106, USA
| | - Jeffrey W Smith
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA
| |
Collapse
|
14
|
Kuwajima K, Chang K, Furuta A, Bougaki M, Uchida K, Sawamura S, Yamada Y. Synergistic cytoprotection by co-treatment with dexamethasone and rapamycin against proinflammatory cytokine-induced alveolar epithelial cell injury. J Intensive Care 2019; 7:12. [PMID: 30774959 PMCID: PMC6367811 DOI: 10.1186/s40560-019-0365-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/22/2019] [Indexed: 11/10/2022] Open
Abstract
Background One of the main pathophysiological manifestations during the acute phase of sepsis is massive production of proinflammatory mediators. Clinical trials involving direct suppression of inflammatory mediators to relieve organ dysfunction in sepsis have been extensively performed; however, the clinical outcomes of such trials remain far from satisfactory. Given the need for better sepsis treatments, we have screened various agents with anti-inflammatory properties for cytoprotective effects. In this study, we identified dexamethasone and rapamycin as clinically applicable candidates with favorable synergistic effects against inflammatory cytokine-induced cytotoxicity in vitro and further explored the molecular mechanisms underlying the augmented cytoprotective effects exerted by co-treatment with both drugs. Methods Human alveolar epithelial cell-derived A549 cells were stimulated with a mixture of inflammatory cytokines, TNF-alpha, IL-1beta, and IFN-gamma, which induce cellular injury, including apoptosis. This in vitro model was designed to simulate acute lung injury (ALI) associated with sepsis. The cells were co-treated with dexamethasone and rapamycin under cytokine stimulation. Conditioned medium and cell lysates were subjected to further analysis. Results Either dexamethasone or rapamycin significantly attenuated cytokine-induced cytotoxicity in A549 cells in a dose-dependent manner. In addition, the simultaneous administration of dexamethasone and rapamycin had a synergistic cytoprotective effect. The applied doses of dexamethasone (10 nM) and rapamycin (1 nM) were considerably below the reported plasma concentrations of each drug in clinical setting. Interestingly, distinct augmentation of both of c-Jun inhibition and Akt activation were observed when the cells were co-treated with both drugs under cytokine stimulation. Conclusions A synergistic protective effect of dexamethasone and rapamycin was observed against cytokine-induced cytotoxicity in A549 cells. Augmentation of both of c-Jun inhibition and Akt activation were likely responsible for the cytoprotective effect. The combined administration of anti-inflammatory drugs such as dexamethasone and rapamycin offers a promising treatment option for alveolar epithelial injury associated with sepsis.
Collapse
Affiliation(s)
- Ken Kuwajima
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kyungho Chang
- 2Anesthesiology and Intensive Care Unit, Teikyo University School of Medicine, Tokyo, Japan
| | - Ai Furuta
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masahiko Bougaki
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shigehito Sawamura
- 2Anesthesiology and Intensive Care Unit, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshitsugu Yamada
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Chen L, Welty-Wolf KE, Kraft BD. Nonhuman primate species as models of human bacterial sepsis. Lab Anim (NY) 2019; 48:57-65. [PMID: 30643274 PMCID: PMC6613635 DOI: 10.1038/s41684-018-0217-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022]
Abstract
Sepsis involves a disordered host response to systemic infection leading to high morbidity and mortality. Despite intense research, targeted sepsis therapies beyond antibiotics have remained elusive. The cornerstone of sepsis research is the development of animal models to mimic human bacterial infections and test novel pharmacologic targets. Nonhuman primates (NHPs) have served as an attractive, but expensive, animal to model human bacterial infections due to their nearly identical cardiopulmonary anatomy and physiology, as well as host response to infection. Several NHP species have provided substantial insight into sepsis-mediated inflammation, endothelial dysfunction, acute lung injury, and multi-organ failure. The use of NHPs has usually focused on translating therapies from early preclinical models to human clinical trials. However, despite successful sepsis interventions in NHP models, there are still no FDA-approved sepsis therapies. This review highlights major NHP models of bacterial sepsis and their relevance to clinical medicine. Treatment for bacterial sepsis remains limited beyond the use of antibiotics. Lingye Chen, Karen Welty-Wolf, and Bryan Kraft review nonhuman primate models of sepsis and highlight their advantages and limitations compared to other preclinical models.
Collapse
Affiliation(s)
- Lingye Chen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Karen E Welty-Wolf
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Bryan D Kraft
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
16
|
Jaffarali M, Hall A. Transcriptional regulation of protein S gene. HAMDAN MEDICAL JOURNAL 2019. [DOI: 10.4103/hmj.hmj_49_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
17
|
Aldag J, Persson T, Hartmann RK. 2'-Fluoro-Pyrimidine-Modified RNA Aptamers Specific for Lipopolysaccharide Binding Protein (LBP). Int J Mol Sci 2018; 19:ijms19123883. [PMID: 30563044 PMCID: PMC6321028 DOI: 10.3390/ijms19123883] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Lipopolysaccaride binding protein (LBP), a glycosylated acute phase protein, plays an important role in the pathophysiology of sepsis. LBP binds with high affinity to the lipid part of bacterial lipopolysaccaride (LPS). Inhibition of the LPS-LBP interaction or blockage of LBP-mediated transfer of LPS monomers to CD14 may be therapeutical strategies to prevent septic shock. LBP is also of interest as a biomarker to identify septic patients at high risk for death, as LBP levels are elevated during early stages of severe sepsis. As a first step toward such potential applications, we isolated aptamers specific for murine LBP (mLBP) by in vitro selection from a library containing a 60-nucleotide randomized region. Modified RNA pools were transcribed in the presence of 2′-fluoro-modified pyrimidine nucleotides to stabilize transcripts against nuclease degradation. As verified for one aptamer experimentally, the selected aptamers adopt a “three-helix junction” architecture, presenting single-stranded 7-nt (5′-YGCTTCY) or 6-nt (5′-RTTTCY) consensus sequences in their core. The best binder (aptamer A011; Kd of 270 nM for binding to mLBP), characterized in more detail by structure probing and boundary analysis, was demonstrated to bind with high specificity to murine LBP.
Collapse
Affiliation(s)
- Jasmin Aldag
- Jasmin Aldag, EUROIMMUN AG, Seekamp 31, D-23560 Lübeck, Germany.
| | - Tina Persson
- Tina Persson, Passage2Pro AB, Östra Kristinelundsvägen 4B, SE-21748 Malmö, Sweden.
| | - Roland K Hartmann
- Roland K. Hartmann, Philipps-Universität Marburg, Institut für Pharmazeutische Chemie, Marbacher Weg 6, D-35037 Marburg, Germany.
| |
Collapse
|
18
|
Onohara T, Sakamoto Y, Inoue S. Plasma Adsorption Membranes Are Able to Efficiently Remove High Mobility Group Box-1 (HMGB-1). J NIPPON MED SCH 2018; 85:150-156. [PMID: 30135341 DOI: 10.1272/jnms.jnms.2018_85-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND High Mobility Group Box 1 (HMGB-1) is a 30 kDa protein that is a lethal mediator in sepsis and is a recognized therapeutic target. However, no consensus has been reached for acute blood purification therapy as a treatment for sepsis targeting HMGB-1. Previous studies demonstrated that a high anti-HMGB-1 antibody titer and the suppression of HMGB-1 activity improved survival rate in animal sepsis models. The aim of this study was to evaluate whether plasma adsorption therapy is able to decrease the level of HMGB-1, representing a new potential treatment strategy against sepsis. METHODS Plasma adsorption therapy has been known as a treatment for various autoimmune diseases. Three different adsorbent columns, including TR-350 (IM-TR), PH-350 (IM-PH), and BRS-350 (BRS), were used in this study for comparison. We made a 1/350 scale of these three columns. Fetal bovine serum (FBS) contains a significant amount of HMGB-1. After priming with saline, FBS was passed through the columns and the adsorption rates of HMGB-1 at 25 minutes, 50 minutes, and 75 minutes were evaluated. The total adsorbed HMGB-1 level at 75 minutes was also calculated. RESULTS The highest adsorption rate and total adsorbed amount of HMGB-1 were observed in IM-TR, followed by BRS and IM-PH. IM-TR showed a decline in adsorption rate over time. BRS showed a steady adsorption rate at all time points. IM-TR removed HMGB-1 significantly more than IM-PH and BRS. CONCLUSIONS Our findings showed that plasma adsorption therapy efficiently adsorbed HMGB-1 and could be safely applied for the treatment of sepsis.
Collapse
Affiliation(s)
- Takayuki Onohara
- The Department of Emergency and Critical Care, Ureshino Medical Center
| | - Yuichiro Sakamoto
- The Department of Emergency and Critical Care Medicine, Saga University Hospital
| | - Satoshi Inoue
- The Division of Trauma Surgery and Surgical Critical Care, Saga University Hospital
| |
Collapse
|
19
|
Zhou Y, Xia Q, Wang X, Fu S. Endotoxin Tolerant Dendritic Cells Suppress Inflammatory Responses in Splenocytes via Interleukin-1 Receptor Associated Kinase (IRAK)-M and Programmed Death-Ligand 1 (PDL-1). Med Sci Monit 2018; 24:4798-4806. [PMID: 29995830 PMCID: PMC6069485 DOI: 10.12659/msm.908242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Sepsis causes the highest mortality in non-cardiovascular intensive care units worldwide. Recent research has demonstrated that the late phase of sepsis, characterized as septic immunosuppression, is the central pathophysiological mechanism of immune dysfunction. Investigating the suppressive mechanism of immune cells may identify possible targets for therapy. MATERIAL AND METHODS We used LPS 2-hit model for dendritic cells (DCs) to establish endotoxin tolerance, and co-cultured with splenocytes. Co-culture responses and gene expressions were evaluated. RESULTS Endotoxin tolerant DCs showed irresponsiveness in pro-inflammatory cytokine production and expressed negative regulator genes of inflammation. When co-cultured with splenocytes, suppression of inflammatory responses and T cells apoptosis were observed with elevated expression of IRAK-M and PDL-1, and interference and neutralization of these 2 molecules led to partly reversed suppression of inflammation. CONCLUSIONS Our research found direct regulation of endotoxin tolerant DCs to other immune cells and suggested a possible mechanism via IRAK-M and PDL-1. This may inform research on septic immunosuppression and suggests possible therapeutic targets for sepsis.
Collapse
Affiliation(s)
- Yuping Zhou
- Department of Anesthesiology, Shanghai Dermatology Hospital, Shanghai, China (mainland)
| | - Qin Xia
- Department of Anesthesiology, Tenth People's Hospital, Tongji University, Shanghai, China (mainland)
| | - Xi Wang
- Department of Anesthesiology, Tenth People's Hospital, Tongji Universit, Shanghai, China (mainland)
| | - Shukun Fu
- Department of Anesthesiology, Tenth People's Hospital, Tongji University, Shanghai, China (mainland)
| |
Collapse
|
20
|
Niu R, Gao H, Zhou Y, Zhang J. Ouabain Attenuates Sepsis-Induced Immunosuppression in Mice by Activation and Anti-Apoptosis of T Cells. Med Sci Monit 2018; 24:2720-2727. [PMID: 29717720 PMCID: PMC5952820 DOI: 10.12659/msm.906889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Sepsis is known to trigger impaired T cell function, which relates to immunosuppression, contributing to refractory infection and high mortality. The mechanisms of T cell recovery remain to be elucidated, and novel and effective therapeutics for sepsis are needed. Ouabain, a small molecule of cardiac glycosides, can reverse immunoparalysis in many settings. Material/Methods Our study was designed to determine if ouabain can relieve sepsis by modulating T cell response and related pathways. The “two-hit” model of sepsis was applied, established by intraperitoneally LPS injection 3 days after cecal ligation puncture (CLP-LPS). Ouabain was administered to mice intravenously (0.1 mg/kg) after in vivo LPS stimulation every day for 4 days. The survival rate of mice, level of serum cytokines, percentage of activated T cells, apoptosis of T cells, and possibly related genes were assessed. Results The results suggest that ouabain administration after establishment of the CLP-LPS model improved survival rates, elevated pro-inflammatory cytokines, and decreased anti-inflammatory cytokines in serum. More activated T cells and fewer apoptotic T cells were detected in the spleens after treatment with ouabain. Such changes might correlate with the genes of Bcl-2, PUMA, IRAK-M, and SOCS1. Conclusions Taken together, our data show ouabain is a T cell mediator during sepsis recovery.
Collapse
Affiliation(s)
- Ruibin Niu
- Department of Anesthesiology, Huashan Hospital North Affiliated to Fudan University, Shanghai, China (mainland)
| | - Hao Gao
- Department of Anesthesiology, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Yuan Zhou
- Department of Anesthesiology, Huashan Hospital North affiliated to Fudan University, Shanghai, China (mainland)
| | - Jie Zhang
- Department of Anesthesiology, Huashan Hospital North affiliated to Fudan University, Shanghai, China (mainland)
| |
Collapse
|
21
|
Clatworthy AE, Romano KP, Hung DT. Whole-organism phenotypic screening for anti-infectives promoting host health. Nat Chem Biol 2018; 14:331-341. [PMID: 29556098 PMCID: PMC9843822 DOI: 10.1038/s41589-018-0018-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/20/2017] [Indexed: 01/19/2023]
Abstract
To date, antibiotics have been identified on the basis of their ability to kill bacteria or inhibit their growth rather than directly for their capacity to improve clinical outcomes of infected patients. Although historically successful, this approach has led to the development of an antibiotic armamentarium that suffers from a number of shortcomings, including the inevitable emergence of resistance and, in certain infections, suboptimal efficacy leading to long treatment durations, infection recurrence, or high mortality and morbidity rates despite apparent bacterial sterilization. Conventional antibiotics fail to address the complexities of in vivo bacterial physiology and virulence, as well as the role of the host underlying the complex, dynamic interactions that cause disease. New interventions are needed, aimed at host outcome rather than microbiological cure. Here we review the role of screening models for cellular and whole-organism infection, including worms, flies, zebrafish, and mice, to identify novel therapeutic strategies and discuss their future implications.
Collapse
Affiliation(s)
- Anne E. Clatworthy
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Keith P. Romano
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Deborah T. Hung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA,Correspondence and requests for materials should be addressed to D.T.H.
| |
Collapse
|
22
|
Burgdorff AM, Bucher M, Schumann J. Vasoplegia in patients with sepsis and septic shock: pathways and mechanisms. J Int Med Res 2018; 46:1303-1310. [PMID: 29332515 PMCID: PMC6091823 DOI: 10.1177/0300060517743836] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sepsis is one of the most frequent causes of death among patients in intensive care units. Many therapeutic strategies have been assessed without the desired success rates. A key risk factor for death is hypotension due to vasodilatation with vascular hyposensitivity. However, the pathways underlying this process remain unclear. Endotoxemia induces inflammatory mediators, and this is followed by vasoplegia and decreased cardiac contractility. Although inhibition of these mediators diminishes mortality rates in animal models, this phenomenon has not been confirmed in humans. Downregulation of vasoconstrictive receptors such as angiotensin receptors, adrenergic and vasopressin receptors is seen in sepsis, which is associated with a hyporesponsiveness to vasoconstrictive mediators. Animal studies have verified that receptor downregulation is linked to the above-mentioned inflammatory mediators. Anti-inflammatory therapy with glucocorticoids reportedly improves responsiveness to catecholamines with higher survival in rats, although this has not been shown to be clinically significant in humans. Hence, there is an urgent need for in-depth studies investigating the underlying mechanisms of vasoplegia to allow for development of effective therapeutic strategies for the treatment of sepsis.
Collapse
Affiliation(s)
- A-M Burgdorff
- Department of Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), Halle (Saale), Germany
| | - M Bucher
- Department of Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), Halle (Saale), Germany
| | - J Schumann
- Department of Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), Halle (Saale), Germany
| |
Collapse
|
23
|
Haase M, Bellomo R, Morger S, Baldwin I, Boyce N. High Cut-off Point Membranes in Septic Acute Renal Failure: A Systematic Review. Int J Artif Organs 2018; 30:1031-41. [DOI: 10.1177/039139880703001202] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objectives To review the literature on the experimental, physiological and clinical effects of blood purification with high cut-off (HCO) point membranes in septic acute renal failure (ARF). Study Design MEDLINE and PubMed database search combining relevant terms and integrating data from studies on the use of HCO membranes. Setting and Population Ex vivo studies of endotoxemia, animal studies of bacteremia and clinical studies using HCO membranes in patients with septic ARF. Selection Criteria for Studies: Original data from primary publications. Interventions: HCO membrane-based hemodialysis, hemodiafiltration or hemofiltration. Outcomes: Plasma cytokine clearance, immunological and physiological effects and safety parameters of HCO membranes. Results HCO membranes effectively remove cytokines from blood. Treatment using HCO membranes has beneficial effects on immune cell function and increases survival in animal models of sepsis. Preliminary clinical studies show that HCO membranes decrease plasma cytokine levels and the need for vasopressor therapy. HCO membrane-based blood purification has now been applied in four pilot randomized controlled studies of 70 patients with septic ARF with no reports of serious adverse effects. Limitations Because of substantial heterogeneity, no formal quantitative analysis could be performed. Conclusions The available evidence on HCO blood purification justifies larger randomized controlled trials in patients with septic ARF.
Collapse
Affiliation(s)
- M. Haase
- Intensive Care Unit, Austin Hospital, University of Melbourne - Australia
- Department of Nephrology and Intensive Care, Charité University Medicine, Berlin - Germany
| | - R. Bellomo
- Intensive Care Unit, Austin Hospital, University of Melbourne - Australia
| | - S. Morger
- Department of Nephrology and Intensive Care, Charité University Medicine, Berlin - Germany
| | - I. Baldwin
- Intensive Care Unit, Austin Hospital, University of Melbourne - Australia
| | - N. Boyce
- Australian Red Cross Blood Service, University of Melbourne - Australia
| |
Collapse
|
24
|
Xia Q, Zhou Y, Wang X, Fu S. Interleukin-1 receptor-associated kinase 3 downregulation in peripheral blood mononuclear cells attenuates immunosuppression in sepsis. Exp Ther Med 2017; 15:1586-1593. [PMID: 29434744 DOI: 10.3892/etm.2017.5549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/13/2017] [Indexed: 12/14/2022] Open
Abstract
Sepsis is the leading cause of mortality in intensive care units due to complex inflammatory immune responses and immunosuppression. Recent studies have indicated that the negative regulator of toll like receptors, interleukin-1 receptor-associated kinase 3 (IRAK-3/IRAK-M), serves an important role in immunosuppression during sepsis. In the current study, a cecal ligation puncture model was established in mice using lipopolysaccharide secondary challenge to simulate immunosuppression in sepsis. Peripheral blood mononuclear cells (PBMCs) from this model were then used to evaluate the expression and function of IRAK-M. The results demonstrated that silencing of IRAK-M expression in PBMCs from immunosuppressed mice partially restored the production of pro-inflammatory cytokines. By introducing PBMCs transfected with small-interfering RNA targeting IRAK-M into septic immunosuppressed mice, the survival rate was improved with an increase in splenic CD4+ and CD8+ T cells and a decrease in T cell apoptosis. In conclusion, downregulation of IRAK-M reversed the effects of sepsis on the production of inflammatory cytokines in PBMCs, and improved the survival of septic immunosuppressed mice. These results provide a basis for future studies investigating the immunological mechanisms underlying immune suppression in sepsis.
Collapse
Affiliation(s)
- Qin Xia
- Department of Anesthesiology, The Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Yupin Zhou
- Department of Anesthesiology, Shanghai Dermatology Hospital, Shanghai 200050, P.R. China
| | - Xi Wang
- Department of Anesthesiology, The Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Shukun Fu
- Department of Anesthesiology, The Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| |
Collapse
|
25
|
Essential roles for platelets during neutrophil-dependent or lymphocyte-mediated defense against bacterial pathogens. Blood Coagul Fibrinolysis 2017; 27:667-72. [PMID: 26588444 DOI: 10.1097/mbc.0000000000000455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Emerging evidence from animal models suggests that platelets may participate in a wide variety of processes including the immune response against infection. More than 200 whole blood samples from patients and healthy controls were run in the System XE-5000 analyzer, and plasma fractions were separated for the following tests by ELISA, Luminex and light scattering. We describe two mechanisms by which platelets may contribute to immune function against various bacterial pathogens based on increased mean platelet volume in gram-positive bacterial infections and increased platelet counts in gram-negative bacterial infections. Gram-negative bacteria activate platelets to recruit neutrophils, which participate in the immune response against infection. During this process, fractalkine, macrophage inflammatory protein-1β, interleukin-17A, tumor necrosis factor-α and platelet-activating factor were higher in patients infected with Escherichia coli; additionally, giant platelets were observed under the microscope. Meanwhile, we found that platelets played a different role in gram-positive bacterial infections. Specifically, they could actively adhere to gram-positive bacteria in circulation and transfer them to immune sites to promote antibacterial lymphocyte expansion. During this process, complement C3 and factor XI were more highly expressed in patients infected with Staphylococcus aureus; additionally, we detected more small platelets under the microscope. Platelets participate in the immune response against both gram-negative and gram-positive bacteria, although the mechanisms differ. These results will help us understand the complex roles of platelets during infections, and direct our use of antibiotics based on clinical platelet data.
Collapse
|
26
|
Li W, Wang X, Niu X, Zhang H, He Z, Wang Y, Zhi W, Liu F. Protective Effects of Nobiletin Against Endotoxic Shock in Mice Through Inhibiting TNF-α, IL-6, and HMGB1 and Regulating NF-κB Pathway. Inflammation 2017; 39:786-97. [PMID: 26846885 DOI: 10.1007/s10753-016-0307-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nobiletin (NOB), the major bioactive component of polymethoxyflavones in citrus fruits, has been reported possessing significant biological properties. The purpose of the present study was to investigate the protective role of NOB on lipopolysaccharide (LPS)-induced endotoxic shock in mice. We found pretreatment with NOB increases the survival rate of mice after endotoxin injection. The present study clearly demonstrates that pretreatment with NOB decreases the production of early pro-inflammatory cytokines TNF-α, IL-6, and late-phase mediator HMGB1 in serum and tissues of kidney, lung, and liver. The histopathological study indicates that NOB administration significantly attenuate tissues injury induced by LPS. Moreover, NOB suppresses the activity of nuclear factor-kappa B (NF-κB). These results suggest that NOB protects mice against LPS-induced endotoxic shock through inhibiting the production of TNF-α, IL-6, and HMGB1 and the activation of NF-κB, which elucidate that NOB may be a promising drug candidate for the treatment of septic shock.
Collapse
Affiliation(s)
- Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| | - Xiumei Wang
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China.
| | - Hailin Zhang
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| | - Zehong He
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| | - Yu Wang
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| | - Wenbing Zhi
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| | - Fang Liu
- School of Pharmacy, Xi'an Jiaotong University, No. 76 Western Yanta Road, Xi'an City, Shaanxi Province, 710061, People's Republic of China
| |
Collapse
|
27
|
|
28
|
Barratt-Due A, Pischke SE, Nilsson PH, Espevik T, Mollnes TE. Dual inhibition of complement and Toll-like receptors as a novel approach to treat inflammatory diseases-C3 or C5 emerge together with CD14 as promising targets. J Leukoc Biol 2016; 101:193-204. [PMID: 27581539 PMCID: PMC5166441 DOI: 10.1189/jlb.3vmr0316-132r] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/01/2016] [Accepted: 07/25/2016] [Indexed: 12/22/2022] Open
Abstract
Review of how targeting key upstream molecules at the recognition phase of innate immunity exert anti-inflammatory effects; a potential therapeutic regimen for inflammatory diseases. The host is protected by pattern recognition systems, including complement and TLRs, which are closely cross-talking. If improperly activated, these systems might induce tissue damage and disease. Inhibition of single downstream proinflammatory cytokines, such as TNF, IL-1β, and IL-6, have failed in clinical sepsis trials, which might not be unexpected, given the substantial amounts of mediators involved in the pathogenesis of this condition. Instead, we have put forward a hypothesis of inhibition at the recognition phase by “dual blockade” of bottleneck molecules of complement and TLRs. By acting upstream and broadly, the dual blockade could be beneficial in conditions with improper or uncontrolled innate immune activation threatening the host. Key bottleneck molecules in these systems that could be targets for inhibition are the central complement molecules C3 and C5 and the important CD14 molecule, which is a coreceptor for several TLRs, including TLR4 and TLR2. This review summarizes current knowledge of inhibition of complement and TLRs alone and in combination, in both sterile and nonsterile inflammatory processes, where activation of these systems is of crucial importance for tissue damage and disease. Thus, dual blockade might provide a general, broad-acting therapeutic regimen against a number of diseases where innate immunity is improperly activated.
Collapse
Affiliation(s)
- Andreas Barratt-Due
- Department of Immunology, Oslo University Hospital, and K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Department of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Søren Erik Pischke
- Department of Immunology, Oslo University Hospital, and K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Department of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital, and K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, and K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway; .,Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Research Laboratory Nordland Hospital, Bodø, Norway; and.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| |
Collapse
|
29
|
Ronagh M, Souder L. The Ethics of Ironic Science in Its Search for Spoof. SCIENCE AND ENGINEERING ETHICS 2015; 21:1537-1549. [PMID: 25510233 DOI: 10.1007/s11948-014-9619-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/09/2014] [Indexed: 06/04/2023]
Abstract
The goal of most scientific research published in peer-review journals is to discover and report the truth. However, the research record includes tongue-in-cheek papers written in the conventional form and style of a research paper. Although these papers were intended to be taken ironically, bibliographic database searches show that many have been subsequently cited as valid research, some in prestigious journals. We attempt to understand why so many readers cited such ironic science seriously. We draw from the literature on error propagation in research publication for ways categorize citations. We adopt the concept of irony from the fields of literary and rhetorical criticism to detect, characterize, and analyze the interpretations in the more than 60 published research papers that cite an instance of ironic science. We find a variety of interpretations: some citing authors interpret the research as valid and accept it, some contradict or reject it, and some acknowledge its ironic nature. We conclude that publishing ironic science in a research journal can lead to the same troubles posed by retracted research, and we recommend relevant changes to publication guidelines.
Collapse
Affiliation(s)
- Maryam Ronagh
- Department of Culture and Communication, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Lawrence Souder
- Department of Culture and Communication, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
30
|
Biron BM, Ayala A, Lomas-Neira JL. Biomarkers for Sepsis: What Is and What Might Be? Biomark Insights 2015; 10:7-17. [PMID: 26417200 PMCID: PMC4571989 DOI: 10.4137/bmi.s29519] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/21/2015] [Accepted: 07/27/2015] [Indexed: 12/29/2022] Open
Abstract
Every year numerous individuals develop the morbid condition of sepsis. Therefore, novel biomarkers that might better inform clinicians treating such patients are sorely needed. Difficulty in identifying such markers is in part due to the complex heterogeneity of sepsis, resulting from the broad and vague definition of this state/condition based on numerous possible clinical signs and symptoms as well as an incomplete understanding of the underlying pathobiology of this complex condition. This review considers some of the attempts that have been made so far, looking at both the pro- and anti-inflammatory response to sepsis, as well as genomic analysis, as sources of potential biomarkers. Irrespective, for functional biomarker(s) of sepsis to successfully translate from the laboratory to a clinical setting, the biomarker must be target specific and sensitive as well as easy to implement/interpret, and be cost effective, such that they can be utilized routinely in patient diagnosis and treatment.
Collapse
Affiliation(s)
- Bethany M Biron
- Division of Surgical Research, Department of Surgery, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Joanne L Lomas-Neira
- Division of Surgical Research, Department of Surgery, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
31
|
Platelets in the immune response: Revisiting platelet-activating factor in anaphylaxis. J Allergy Clin Immunol 2015; 135:1424-32. [PMID: 26051949 DOI: 10.1016/j.jaci.2015.04.019] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/21/2015] [Accepted: 04/21/2015] [Indexed: 01/09/2023]
Abstract
Anaphylaxis is an acute, severe, life-threatening multisystem allergic reaction resulting from the sudden systemic release of biochemical mediators and chemotactic substances. Release of both preformed granule-associated mediators and newly generated lipid-derived mediators contributes to the amplification and prolongation of anaphylaxis. Platelet-activating factor (PAF) is a potent phospholipid-derived mediator the central role of which has been well established in experimental models of both immune-mediated and non-immune mediated anaphylaxis. It is produced and secreted by several types of cells, including mast cells, monocytes, tissue macrophages, platelets, eosinophils, endothelial cells, and neutrophils. PAF is implicated in platelet aggregation and activation through release of vasoactive amines in the inflammatory response, resulting in increased vascular permeability, circulatory collapse, decreased cardiac output, and various other biological effects. PAF is rapidly hydrolyzed and degraded to an inactive metabolite, lysoPAF, by the enzyme PAF acetylhydrolase, the activity of which has shown to correlate inversely with PAF levels and predispose to severe anaphylaxis. In addition to its role in anaphylaxis, PAF has also been implicated as a mediator in both allergic and nonallergic inflammatory diseases, including allergic rhinitis, sepsis, atherosclerotic disease, and malignancy, in which PAF signaling has an established role. The therapeutic role of PAF antagonism has been investigated for several diseases, with variable results thus far. Further investigation of its role in pathology and therapeutic modulation is highly anticipated because of the pressing need for more selective and targeted therapy for the management of severe anaphylaxis.
Collapse
|
32
|
Ulland TK, Ferguson PJ, Sutterwala FS. Evasion of inflammasome activation by microbial pathogens. J Clin Invest 2015; 125:469-77. [PMID: 25642707 DOI: 10.1172/jci75254] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Activation of the inflammasome occurs in response to infection with a wide array of pathogenic microbes. The inflammasome serves as a platform to activate caspase-1, which results in the subsequent processing and secretion of the proinflammatory cytokines IL-1β and IL-18 and the initiation of an inflammatory cell death pathway termed pyroptosis. Effective inflammasome activation is essential in controlling pathogen replication as well as initiating adaptive immune responses against the offending pathogens. However, a number of pathogens have developed strategies to evade inflammasome activation. In this Review, we discuss these pathogen evasion strategies as well as the potential infectious complications of therapeutic blockade of IL-1 pathways.
Collapse
|
33
|
Shankar-Hari M, Donnelly A, Pinto R, Salih Z, McKenzie C, Terblanche M, Adhikari NKJ. The influence of statin exposure on inflammatory markers in patients with early bacterial infection: pilot prospective cohort study. BMC Anesthesiol 2014; 14:106. [PMID: 25484622 PMCID: PMC4256798 DOI: 10.1186/1471-2253-14-106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 09/25/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In the context of infection, progressive illness resulting in acute organ dysfunction is thought to be secondary to inflammatory response. Our aims were to determine risk factors for progressive illness following infection in a low-risk hospitalised cohort, including the impact of prior stain therapy. METHODS We performed a prospective observational cohort study on two adult acute medical wards of a single tertiary academic hospital. We screened drug prescription charts of all adult acute medical admissions for inclusion criteria of inpatient administration of antibiotics for more than 24 hours for a microbiologically confirmed or clinically suspected infection. Patients were followed until admission to a high dependency unit (HDU) or intensive care unit (ICU), discharge from hospital, or to a maximum of 10 days. Outcomes were evolution of systemic inflammatory response syndrome (SIRS) criteria, white cell count and C-reactive protein measurements, and adverse clinical outcomes. We constructed multivariable models accounting for repeated within-patient measurements to determine associations between a priori selected predictors (days since admission, age, gender, Charlson score, prior statin exposure) and selected outcomes. RESULTS We enrolled 209 patients; 27.8% were statin users and the commonest infection was pneumonia (51.0%). Most (88.0%) had at least 1 SIRS criterion on admission, and 76 (37.3%) manifested additional SIRS criteria over time. Risks of admission to HDU/ICU (3.3%) and of 30-day mortality (5.7%) were low. The proportion of patients with at least 1 SIRS criterion, mean CRP, and mean WBC all decreased over time. Multivariable regression models identified days since hospital admission as the only variable associated with daily presence of SIRS criteria, WCC, or CRP (adjusted OR <1 and p < 0.0001 in all analyses). Statin exposure was not a significant predictor. CONCLUSIONS This cohort of ward patients treated for infection had a low risk of clinical deterioration, inflammatory markers improved over time, and statin exposure was not associated with any outcome. Future larger studies may identify risk factors for progression of illness in this population and plausible surrogate endpoints for evaluation in clinical trials.
Collapse
Affiliation(s)
- Manu Shankar-Hari
- Division of Asthma, Allergy and Lung Biology, King's College London, Department of Critical Care Medicine, St. Thomas' Hospital, 1st Floor, East Wing, Westminster Bridge Road, London, SE1 7EH UK
| | - Antonia Donnelly
- Emergency Department, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, SO16 6YD UK
| | - Ruxandra Pinto
- Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, M4N 3M5 Canada
| | - Zaid Salih
- School of Pharmacy, University College London, London, UK ; Department of Pharmacy and Critical Care, Guys and St Thomas NHS Foundation Trust, St Thomas' Hospital, Lambeth Palace Road, London, SE1 7EH UK
| | - Cathrine McKenzie
- Department of Pharmacy and Critical Care, Guys and St Thomas NHS Foundation Trust, St Thomas' Hospital, Lambeth Palace Road, London, SE1 7EH UK ; Institute of Pharmaceutical Sciences, Kings College London, London, UK
| | - Marius Terblanche
- Centre for Critical Illness Data Sciences Research, Division of Health & Social Care Research, King's College London, Department of Critical Care Medicine, 1st Floor, East Wing, St. Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH UK
| | - Neill K J Adhikari
- Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute and University of Toronto, 2075 Bayview Avenue, Toronto, M4N 3M5 Canada
| |
Collapse
|
34
|
Chen Y, Zhang P, Xu SC, Yang L, Voss U, Ekblad E, Wu Y, Min Y, Hertervig E, Nilsson Å, Duan RD. Enhanced colonic tumorigenesis in alkaline sphingomyelinase (NPP7) knockout mice. Mol Cancer Ther 2014; 14:259-67. [PMID: 25381265 DOI: 10.1158/1535-7163.mct-14-0468-t] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intestinal alkaline sphingomyelinase (alk-SMase) generates ceramide and inactivates platelet-activating factor (PAF) and was previously suggested to have anticancer properties. The direct evidence is still lacking. We studied colonic tumorigenesis in alk-SMase knockout (KO) mice. Formation of aberrant crypt foci (ACF) was examined after azoxymethane (AOM) injection. Tumor was induced by AOM alone, a conventional AOM/dextran sulfate sodium (DSS) treatment, and an enhanced AOM/DSS method. β-Catenin was determined by immunohistochemistry, PAF levels by ELISA, and sphingomyelin metabolites by mass spectrometry. Without treatment, spontaneous tumorigenesis was not identified but the intestinal mucosa appeared thicker in KO than in wild-type (WT) littermates. AOM alone induced more ACF in KO mice but no tumors 28 weeks after injection. However, combination of AOM/DSS treatments induced colonic tumors and the incidence was significantly higher in KO than in WT mice. By the enhanced AOM/DSS method, tumor number per mouse increased 4.5 times and tumor size 1.8 times in KO compared with WT mice. Although all tumors were adenomas in WT mice, 32% were adenocarcinomas in KO mice. Compared with WT mice, cytosol expression of β-catenin was significantly increased and nuclear translocation in tumors was more pronounced in KO mice. Lipid analysis showed decreased ceramide in small intestine and increased sphingosine-1-phosphate (S1P) in both small intestine and colon in nontreated KO mice. PAF levels in feces were significantly higher in the KO mice after AOM/DSS treatment. In conclusion, lack of alk-SMase markedly increases AOM/DSS-induced colonic tumorigenesis associated with decreased ceramide and increased S1P and PAF levels.
Collapse
Affiliation(s)
- Ying Chen
- Gastroenterology and Nutrition Laboratory, Department of Clinical Sciences Lund, University of Lund, Lund, Sweden. Gastroenterology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Zhang
- Gastroenterology and Nutrition Laboratory, Department of Clinical Sciences Lund, University of Lund, Lund, Sweden
| | - Shu-Chang Xu
- Gastroenterology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Liping Yang
- Cancer Research Center, Tumor Hospital of Nantong University, Nantong, China
| | - Ulrikke Voss
- Neurogastroenterology, Department of Experimental Medical Science, University of Lund, Lund, Sweden
| | - Eva Ekblad
- Neurogastroenterology, Department of Experimental Medical Science, University of Lund, Lund, Sweden
| | - Yunjin Wu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yalan Min
- Cancer Research Center, Tumor Hospital of Nantong University, Nantong, China
| | - Erik Hertervig
- Gastroenterology and Nutrition Laboratory, Department of Clinical Sciences Lund, University of Lund, Lund, Sweden. Gastroenterology, Skåne University Hospital, Lund, Sweden
| | - Åke Nilsson
- Gastroenterology and Nutrition Laboratory, Department of Clinical Sciences Lund, University of Lund, Lund, Sweden. Gastroenterology, Skåne University Hospital, Lund, Sweden
| | - Rui-Dong Duan
- Gastroenterology and Nutrition Laboratory, Department of Clinical Sciences Lund, University of Lund, Lund, Sweden.
| |
Collapse
|
35
|
Cuello F, Shankar-Hari M, Mayr U, Yin X, Marshall M, Suna G, Willeit P, Langley SR, Jayawardhana T, Zeller T, Terblanche M, Shah AM, Mayr M. Redox state of pentraxin 3 as a novel biomarker for resolution of inflammation and survival in sepsis. Mol Cell Proteomics 2014; 13:2545-57. [PMID: 24958171 PMCID: PMC4188985 DOI: 10.1074/mcp.m114.039446] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In an endotoxaemic mouse model of sepsis, a tissue-based proteomics approach for biomarker discovery identified long pentraxin 3 (PTX3) as the lead candidate for inflamed myocardium. When the redox-sensitive oligomerization state of PTX3 was further investigated, PTX3 accumulated as an octamer as a result of disulfide-bond formation in heart, kidney, and lung—common organ dysfunctions seen in patients with sepsis. Oligomeric moieties of PTX3 were also detectable in circulation. The oligomerization state of PTX3 was quantified over the first 11 days in critically ill adult patients with sepsis. On admission day, there was no difference in the oligomerization state of PTX3 between survivors and non-survivors. From day 2 onward, the conversion of octameric to monomeric PTX3 was consistently associated with a greater survival after 28 days of follow-up. For example, by day 2 post-admission, octameric PTX3 was barely detectable in survivors, but it still constituted more than half of the total PTX3 in non-survivors (p < 0.001). Monomeric PTX3 was inversely associated with cardiac damage markers NT-proBNP and high-sensitivity troponin I and T. Relative to the conventional measurements of total PTX3 or NT-proBNP, the oligomerization of PTX3 was a superior predictor of disease outcome.
Collapse
Affiliation(s)
- Friederike Cuello
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK; §Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Centre, University Medical Center Hamburg-Eppendorf, Hamburg, 20246 Germany; ¶DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Manu Shankar-Hari
- ‖Critical Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH UK; **Division of Asthma Allergy and Lung Biology, King's College, London SE1 9RT, UK
| | - Ursula Mayr
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Xiaoke Yin
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Melanie Marshall
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Gonca Suna
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Peter Willeit
- ‡‡Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; §§Department of Neurology, Innsbruck Medical University, Innsbruck, 6020 Austria
| | - Sarah R Langley
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Tamani Jayawardhana
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Tanja Zeller
- ¶¶Clinic for General and Interventional Cardiology, University Heart Centre Hamburg, Hamburg 20246, Germany
| | - Marius Terblanche
- ‖Critical Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH UK
| | - Ajay M Shah
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Manuel Mayr
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK;
| |
Collapse
|
36
|
Esper SA, Subramaniam K, Tanaka KA. Pathophysiology of Cardiopulmonary Bypass. Semin Cardiothorac Vasc Anesth 2014; 18:161-76. [DOI: 10.1177/1089253214532375] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The techniques and equipment of cardiopulmonary bypass (CPB) have evolved over the past 60 years, and numerous numbers of cardiac surgical procedures are conducted around the world using CPB. Despite more widespread applications of percutaneous coronary and valvular interventions, the need for cardiac surgery using CPB remains the standard approach for certain cardiac pathologies because some patients are ineligible for percutaneous procedures, or such procedures are unsuccessful in some. The ageing patient population for cardiac surgery poses a number of clinical challenges, including anemia, decreased cardiopulmonary reserve, chronic antithrombotic therapy, neurocognitive dysfunction, and renal insufficiency. The use of CPB is associated with inductions of systemic inflammatory responses involving both cellular and humoral interactions. Inflammatory pathways are complex and redundant, and thus, the reactions can be profoundly amplified to produce a multiorgan dysfunction that can manifest as capillary leak syndrome, coagulopathy, respiratory failure, myocardial dysfunction, renal insufficiency, and neurocognitive decline. In this review, pathophysiological aspects of CPB are considered from a practical point of view, and preventive strategies for hemodilutional anemia, coagulopathy, inflammation, metabolic derangement, and neurocognitive and renal dysfunction are discussed.
Collapse
|
37
|
Shrum B, Anantha RV, Xu SX, Donnelly M, Haeryfar SMM, McCormick JK, Mele T. A robust scoring system to evaluate sepsis severity in an animal model. BMC Res Notes 2014; 7:233. [PMID: 24725742 PMCID: PMC4022086 DOI: 10.1186/1756-0500-7-233] [Citation(s) in RCA: 283] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/09/2014] [Indexed: 12/19/2022] Open
Abstract
Background The lack of a reliable scoring system that predicts the development of septic shock and death precludes comparison of disease and/or treatment outcomes in animal models of sepsis. We developed a murine sepsis score (MSS) that evaluates seven clinical variables, and sought to assess its validity and reliability in an experimental mouse model of polymicrobial sepsis. Methods Stool collected from the cecum of C57BL/6 (B6) mice was dissolved in 0.9% normal saline (NS) and filtered, resulting in a fecal solution (FS) which was injected intraperitoneally into B6 mice. Disease severity was monitored by MSS during the experimental timeline. Blood and tissue samples were harvested for the evaluation of inflammatory changes after sepsis induction. The correlation between pro-inflammatory markers and MSS was assessed by the Spearman rank correlation coefficient. Results Mice injected with FS at a concentration of 90 mg/mL developed polymicrobial sepsis with a 75% mortality rate at 24 hours. The MSS was highly predictive of sepsis progression and mortality, with excellent discriminatory power, high internal consistency (Cronbach alpha coefficient = 0.92), and excellent inter-rater reliability (intra-class coefficient = 0.96). An MSS of 3 had a specificity of 100% for predicting onset of septic shock and death within 24 hours. Hepatic dysfunction and systemic pro-inflammatory responses were confirmed by biochemical and cytokine analyses where the latter correlated well with the MSS. Significant bacterial dissemination was noted in multiple organs. Furthermore, the liver, spleen, and intestine demonstrated histopathological evidence of injury. Conclusions The MSS reliably predicts disease progression and mortality in an animal model of polymicrobial sepsis. More importantly, it may be used to assess and compare outcomes among various experimental models of sepsis, and serve as an ethically acceptable alternative to death as an endpoint.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tina Mele
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
38
|
Inhibition of sepsis-induced inflammatory response by β1-adrenergic antagonists. J Trauma Acute Care Surg 2014; 76:320-7; discussion 327-8. [PMID: 24458040 DOI: 10.1097/ta.0000000000000113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Although previous studies have described potential benefits of nonselective β-adrenergic antagonist therapy in sepsis, there is a paucity of data on the use of β1-selective antagonists (B1AA). The purposes of this study were to describe the effects of B1AA on survival in septic animals and to explore for molecular mechanisms of potential treatment benefit. METHODS C57BL/6 male mice received intraperitoneal injection of lipopolysaccharide. Continuous infusion of a B1AA (esmolol) or an equal volume of saline (control) was initiated at 4 hours after injection. Kaplan-Meier survival analysis at 120 hours was used to explore for mortality differences. A subgroup of animals was sacrificed for microarray expression analysis. Top candidate genes were validated in vitro and in silico. Expression of our candidate genes in a human microarray database (GSE28750) was explored. RESULTS B1AA infusion resulted in increased survival (p = 0.001) at 120 hours. Mean survival difference was 23.6 hours (p = 0.002). Hazard ratio for mortality with B1AA is 0.43 (95% confidence interval, 0.26-0.72). Immunologic disease (p = 0.0003-0.036) and cell death/survival (p = 0.0001-0.042) were significantly associated with improved survival in septic mice treated with B1AA. Further analysis of the gene structure revealed that eight genes shared common promoter activating sequence for NFKB and/or BRCA1 motifs. Analysis of a human sepsis database identified the up-regulation of CAMP (p = 0.032) and TNFSF10 (p = 0.001) genes in septic patients compared with healthy controls. CONCLUSION Continuous infusion of a B1AA initiated after septic insult improves survival at 5 days in a murine model. Benefits may be caused by modulation of gene expression in immunologic pathways leading to an increase in CAMP and TNFSF10 expression. This observed effect may be explained by the activation of NFKB and BRCA1 genes involved in immune response and cell repair pathways. Our findings support further investigation of the use of B1AA in the treatment of sepsis.
Collapse
|
39
|
Shankar-Hari M, Lord GM. How might a diagnostic microRNA signature be used to speed up the diagnosis of sepsis? Expert Rev Mol Diagn 2014; 14:249-51. [DOI: 10.1586/14737159.2014.899151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Jia SH, Parodo J, Charbonney E, Tsang JLY, Jia SY, Rotstein OD, Kapus A, Marshall JC. Activated neutrophils induce epithelial cell apoptosis through oxidant-dependent tyrosine dephosphorylation of caspase-8. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1030-1040. [PMID: 24589337 DOI: 10.1016/j.ajpath.2013.12.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 11/19/2013] [Accepted: 12/16/2013] [Indexed: 01/12/2023]
Abstract
Activated neutrophils can injure host cells through direct effects of oxidants on membrane phospholipids, but an ability to induce apoptotic cell death has not previously been reported. We show that neutrophils activated in vivo in patients who have sustained multiple trauma or in vitro by exposure to bacterial lipopolysaccharide promote epithelial cell apoptosis through SHP-1-mediated dephosphorylation of epithelial cell caspase-8. Epithelial cell apoptosis induced by circulating neutrophils from patients who had sustained serious injury depended on the generation of neutrophil-derived reactive oxygen intermediates and was blocked by inhibition of NADPH oxidase or restoration of intracellular glutathione. Caspase-8 was constitutively tyrosine phosphorylated in a panel of resting epithelial cells, but underwent SHP-1-dependent dephosphorylation in response to hydrogen peroxide, activated neutrophils, or inhibition of Src kinases. Cells transfected with a mutant caspase-8 in which tyrosine residues at Tyr397 or Tyr465 are replaced by nonphosphorylatable phenylalanine underwent accelerated apoptosis, whereas either mutation of these residues to phosphomimetic glutamic acid or transfection with the Src kinases Lyn or c-Src inhibited hydrogen peroxide-induced apoptosis. Exposure to either hydrogen peroxide or lipopolysaccharide-stimulated neutrophils increased phosphorylation and activity of the phosphatase SHP-1, increased activity of caspases 8 and 3, and accelerated epithelial cell apoptosis. These observations reveal a novel mechanism for neutrophil-mediated tissue injury through oxidant-dependent, SHP-1-mediated dephosphorylation of caspase-8 resulting in enhanced epithelial cell apoptosis.
Collapse
Affiliation(s)
- Song Hui Jia
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada
| | - Jean Parodo
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada
| | - Emmanuel Charbonney
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada; Department of Critical Care Medicine, St. Michael's Hospital, Tortonto, Ontario, Canada
| | - Jennifer L Y Tsang
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada; Department of Critical Care Medicine, St. Michael's Hospital, Tortonto, Ontario, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sang Yang Jia
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada
| | - Ori D Rotstein
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada; Department of Surgery, St. Michael's Hospital, Tortonto, Ontario, Canada
| | - Andras Kapus
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada; Department of Surgery, St. Michael's Hospital, Tortonto, Ontario, Canada
| | - John C Marshall
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, Tortonto, Ontario, Canada; Department of Critical Care Medicine, St. Michael's Hospital, Tortonto, Ontario, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, St. Michael's Hospital, Tortonto, Ontario, Canada.
| |
Collapse
|
41
|
Why have clinical trials in sepsis failed? Trends Mol Med 2014; 20:195-203. [PMID: 24581450 DOI: 10.1016/j.molmed.2014.01.007] [Citation(s) in RCA: 497] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 01/16/2014] [Accepted: 01/22/2014] [Indexed: 12/17/2022]
Abstract
The systemic inflammatory response is biologically complex, redundant, and activated by both infectious and noninfectious triggers. Its manipulation can cause both benefit and harm. More than 100 randomized clinical trials have tested the hypothesis that modulating the septic response to infection can improve survival. With one short-lived exception, none of these has resulted in new treatments. The current challenge for sepsis research lies in a failure of concept and reluctance to abandon a demonstrably ineffectual research model. Future success will necessitate large studies of clinical and biochemical epidemiology to understand the course of illness, better integration of basic and clinical science, and the creation of stratification systems to target treatment towards those who are most likely to benefit.
Collapse
|
42
|
Kosovrasti VY, Lukashev D, Nechev LV, Amiji MM. Novel RNA interference-based therapies for sepsis. Expert Opin Biol Ther 2014; 14:419-35. [PMID: 24397825 DOI: 10.1517/14712598.2014.875524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Sepsis is an extremely fast-paced disease, initiated by an infection that can progress to multiple organ dysfunction and death. The complexity associated with sepsis makes the therapies difficult to develop. Moreover, the 'one-fits-all' kind of therapy is far from being realistic. AREAS COVERED This review provides a conspectus of the current results of sepsis therapies and their benefits, focusing on the development of small interfering RNA (siRNA) therapeutics for targeting immune cells and sepsis pathways. EXPERT OPINION The question, 'When will an effective therapy for sepsis be available for patients?' remains unanswered. New RNA interference-mediated therapies are emerging as novel approaches for the treatment of sepsis by downregulating key inflammatory cytokine expression. Strategies that exploit multimodal gene silencing using siRNA and targeted delivery systems are discussed in this review. Some of these strategies have shown positive results in preclinical model of sepsis.
Collapse
Affiliation(s)
- Verbena Y Kosovrasti
- Northeastern University, School of Pharmacy, Department of Pharmaceutical Sciences , 140 The Fenway Building, R170, 360 Huntington Avenue, Boston, MA 02115 , USA
| | | | | | | |
Collapse
|
43
|
Antitumor necrosis factor therapy is associated with improved survival in clinical sepsis trials: a meta-analysis. Crit Care Med 2013; 41:2419-29. [PMID: 23887234 DOI: 10.1097/ccm.0b013e3182982add] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Sepsis is a lethal syndrome annually affecting approximately 900,000 patients in the United States alone. Despite their benefit in rheumatoid disease, selective antitumor necrosis factor agents failed to improve outcome in early sepsis trials in the 1990s. However, data from additional sepsis trials testing these agents are now available. We therefore sought to determine the effect on survival of selective antitumor necrosis factor agents in randomized clinical sepsis trials.. DATA SOURCES PubMed, Scopus, Embase, and Web of Science. STUDY SELECTION Randomized human sepsis trials of selective antitumor necrosis factor agents reporting survival rates. DATA EXTRACTION Two investigators independently collected relevant data on study characteristics, treatment interventions, and patients from each study. DATA SYNTHESIS Antitumor necrosis factor agents in 15 sepsis trials (n=8,896 patients) meeting inclusion criteria had similar effects (I=0, p=0.84) and compared with controls (placebo in 14 trials or a lower dose in one trial) overall decreased the relative risk of death (95% CI) (0.93 [0.88-0.98], p=0.01). In subgroup analysis, tumor necrosis factor monoclonal antibodies (10 trials, n=6,818) alone produced a significant survival benefit (0.93 [0.87, 0.99], p=0.02) (I=0, p=0.83). Tumor necrosis factor polyclonal antibodies (two trials, n=151) and low-molecular-weight soluble receptor (two trials, n=1,786) had similar beneficial effects to antitumor necrosis factor agents overall (0.82 [0.49-1.37], p=0.45; 0.93 [0.81-1.08], p=0.33, respectively). The effect of tumor necrosis factor high-molecular-weight soluble receptor (one trial, n=141) was not significantly different from other agents but was on the side of harm (1.50 [0.86-2.61], p=0.16). CONCLUSION Antitumor necrosis factor agents produced a modest but significant decrease in the risk of dying with sepsis. Prior individual trials failed to demonstrate benefit, likely because they were underpowered. A definitive trial demonstrating the potential benefit of such agents might require 10,000 or more patients with sepsis.
Collapse
|
44
|
Teixeira-da-Cunha MGA, Gomes RN, Roehrs N, Bozza FA, Prescott SM, Stafforini D, Zimmerman GA, Bozza PT, Castro-Faria-Neto HC. Bacterial clearance is improved in septic mice by platelet-activating factor-acetylhydrolase (PAF-AH) administration. PLoS One 2013; 8:e74567. [PMID: 24069320 PMCID: PMC3771912 DOI: 10.1371/journal.pone.0074567] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 08/05/2013] [Indexed: 11/18/2022] Open
Abstract
Current evidence indicates that dysregulation of the host inflammatory response to infectious agents is central to the mortality of patients with sepsis. Strategies to block inflammatory mediators such as PAF have been investigated as adjuvant therapies for sepsis. PAF-AH, the enzyme responsible for PAF degradation, showed positive results in pre-clinical studies and phase II clinical trials, but the results of a phase III study were disappointing. In this study, we investigated the potential protective mechanism of PAF-AH in sepsis using the murine model of cecal ligation and puncture (CLP). Treatment with rPAF-AH increased peritoneal fluid levels of the anti-inflammatory mediators MCP-1/CCL2 after CLP. The numbers of bacteria (CFU) in the peritoneal cavity were decreased in the rPAF-AH-treated group, indicating more efficient bacterial clearance after rPAF-AH treatment. Interestingly, we observed increased levels of nitric oxide (NO) after PAF-AH administration, and rPAF-AH treatment did not decrease CFU numbers either in iNOS-deficient mice or in CCR2-deficient mice. We concluded that administration of exogenous rPAF-AH reduced inflammatory injury, altered cytokine levels and favored bacterial clearance with a clear impact on mortality through modulation of MCP-1/CCL2 and NO levels in a clinically relevant sepsis model.
Collapse
Affiliation(s)
| | - Rachel N. Gomes
- Laboratório De Imunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, RJ, Brazil
- Laboratório de Investigação em Medicina Intensiva, IPEC, Fiocruz, RJ, Brazil
- * E-mail:
| | - Nathassia Roehrs
- Laboratório De Imunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, RJ, Brazil
| | - Fernando A. Bozza
- Laboratório de Investigação em Medicina Intensiva, IPEC, Fiocruz, RJ, Brazil
| | - Stephen M. Prescott
- Oklahoma Medical Research Foundation, Oklahoma City, United States of America
| | - Diana Stafforini
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
| | - Guy A. Zimmerman
- Program in Human Molecular Biology and Genetics, University of Utah, Salt Lake City, Utah, United States of America
| | - Patricia T. Bozza
- Laboratório De Imunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, RJ, Brazil
| | | |
Collapse
|
45
|
Shedding of tumor necrosis factor receptor 1 induced by protein A decreases tumor necrosis factor alpha availability and inflammation during systemic Staphylococcus aureus infection. Infect Immun 2013; 81:4200-7. [PMID: 24002060 DOI: 10.1128/iai.00593-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus infections are an important public health concern due to their increasing incidence and high rates of mortality. The success of S. aureus as a pathogen is highly related to its enormous capacity to evade the host immune response. The critical role of tumor necrosis factor alpha (TNF-α) in the initial host defense against systemic staphylococcal infection has been demonstrated in experimental models and may partially explain the lack of significant benefits observed in clinical trials attempting to neutralize this cytokine in septic patients. S. aureus protein A plays a key role in regulating inflammation through its ability to bind and signal through the TNF-α receptor 1 (TNFR1). In this study, we demonstrate that S. aureus, via protein A-mediated signaling, induces early shedding of TNFR1, which precedes the secretion of TNF-α in vitro and in vivo. The results obtained using a protein A-deficient mutant and tnfr1(-/-) mice strongly suggest that the increased levels of soluble TNFR1 present during experimental S. aureus infection may neutralize circulating TNF-α and impair the host inflammatory response. Early shedding of TNFR1 induced by protein A may constitute a novel mechanism by which S. aureus subverts the host immune response.
Collapse
|
46
|
Langley RJ, Tsalik EL, van Velkinburgh JC, Glickman SW, Rice BJ, Wang C, Chen B, Carin L, Suarez A, Mohney RP, Freeman DH, Wang M, You J, Wulff J, Thompson JW, Moseley MA, Reisinger S, Edmonds BT, Grinnell B, Nelson DR, Dinwiddie DL, Miller NA, Saunders CJ, Soden SS, Rogers AJ, Gazourian L, Fredenburgh LE, Massaro AF, Baron RM, Choi AMK, Corey GR, Ginsburg GS, Cairns CB, Otero RM, Fowler VG, Rivers EP, Woods CW, Kingsmore SF. An integrated clinico-metabolomic model improves prediction of death in sepsis. Sci Transl Med 2013; 5:195ra95. [PMID: 23884467 DOI: 10.1126/scitranslmed.3005893] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sepsis is a common cause of death, but outcomes in individual patients are difficult to predict. Elucidating the molecular processes that differ between sepsis patients who survive and those who die may permit more appropriate treatments to be deployed. We examined the clinical features and the plasma metabolome and proteome of patients with and without community-acquired sepsis, upon their arrival at hospital emergency departments and 24 hours later. The metabolomes and proteomes of patients at hospital admittance who would ultimately die differed markedly from those of patients who would survive. The different profiles of proteins and metabolites clustered into the following groups: fatty acid transport and β-oxidation, gluconeogenesis, and the citric acid cycle. They differed consistently among several sets of patients, and diverged more as death approached. In contrast, the metabolomes and proteomes of surviving patients with mild sepsis did not differ from survivors with severe sepsis or septic shock. An algorithm derived from clinical features together with measurements of five metabolites predicted patient survival. This algorithm may help to guide the treatment of individual patients with sepsis.
Collapse
|
47
|
Development of a genomic metric that can be rapidly used to predict clinical outcome in severely injured trauma patients. Crit Care Med 2013; 41:1175-85. [PMID: 23388514 DOI: 10.1097/ccm.0b013e318277131c] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Many patients have complicated recoveries following severe trauma due to the development of organ injury. Physiological and anatomical prognosticators have had limited success in predicting clinical trajectories. We report on the development and retrospective validation of a simple genomic composite score that can be rapidly used to predict clinical outcomes. DESIGN Retrospective cohort study. SETTING Multi-institutional level 1 trauma centers. PATIENTS Data were collected from 167 severely traumatized (injury severity score >15) adult (18-55 yr) patients. METHODS Microarray-derived genomic data obtained from 167 severely traumatized patients over 28 days were assessed for differences in messenger RNA abundance among individuals with different clinical trajectories. Once a set of genes was identified based on differences in expression over the entire study period, messenger RNA abundance from these subjects obtained in the first 24 hours was analyzed in a blinded fashion using a rapid multiplex platform, and genomic data reduced to a single metric. RESULTS From the existing genomic dataset, we identified 63 genes whose leukocyte expression differed between an uncomplicated and complicated clinical outcome over 28 days. Using a multiplex approach that can quantitate messenger RNA abundance in less than 12 hours, we reassessed total messenger RNA abundance from the first 24 hours after trauma and reduced the genomic data to a single composite score using the difference from reference. This composite score showed good discriminatory capacity to distinguish patients with a complicated outcome (area under a receiver-operator curve, 0.811; p <0.001). This was significantly better than the predictive power of either Acute Physiology and Chronic Health Evaluation II or new injury severity score scoring systems. CONCLUSIONS A rapid genomic composite score obtained in the first 24 hours after trauma can retrospectively identify trauma patients who are likely to develop complicated clinical trajectories. A novel platform is described in which this genomic score can be obtained within 12 hours of blood collection, making it available for clinical decision making.
Collapse
|
48
|
Abstract
Bacterial clearance is one of the most important beneficial consequences of the innate immune response. Chemokines are important mediators controlling leukocyte trafficking and activation, whereas reactive oxygen and nitrogen species are effectors in bacterial killing. In the present work, we used in vivo and in vitro models of infections to study the role of monocyte chemoattractant protein 1 (MCP-1)/CCL2 and nitric oxide (NO) in the bacterial clearance in sepsis. Our results show that MCP-1/CCL2 and NO levels are increased in the peritoneal cavity of mice 6 h after sepsis induced by cecal ligation and puncture. Pretreatment with anti-MCP-1/CCL2 monoclonal antibodies increased the number of colony-forming units (CFUs) recovered in the peritoneal lavage fluid. Moreover, CFU counts were increased in the peritoneal fluid of CCR2 mice subjected to cecal ligation and puncture. In vitro stimulation of peritoneal macrophages with recombinant MCP-1/CCL2 reduced CFU counts in the supernatant after challenge with Escherichia coli. Conversely, treatment with anti-MCP-1/CCL2 increased CFU counts under the same experimental condition. Stimulation of cultured macrophages with MCP-1/CCL2 and interferon had a synergistic effect on NO production. Macrophages from CCL2 mice showed a consistent decrease in NO production when compared with wild-type controls after stimulation with LPS + interferon. Finally, we showed incubation of macrophages with E. coli, and the ERK inhibitor U0126 increased CFU numbers and decreased intracellular levels of NO. In conclusion, we demonstrated for the first time that MCP-1/CCL2 has a crucial role in the clearance of bacteria by mechanisms involving increased expression of inducible NO synthase and production of NO by ERK signaling pathways.
Collapse
|
49
|
Gentile LF, Cuenca AG, Vanzant EL, Efron PA, McKinley B, Moore F, Moldawer LL. Is there value in plasma cytokine measurements in patients with severe trauma and sepsis? Methods 2013; 61:3-9. [PMID: 23669589 DOI: 10.1016/j.ymeth.2013.04.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 04/18/2013] [Accepted: 04/30/2013] [Indexed: 12/29/2022] Open
Abstract
For the past thirty years, since IL-1β and TNFα were first cloned, there have been efforts to measure plasma cytokine concentrations in patients with severe sepsis and trauma, and to use these measurements to predict clinical outcome and response to therapies. The numbers of cytokines and chemokines that have been measured in the plasma have literally exploded with the development of multiplex immune approaches. Dozens of relatively small cohort studies have shown plasma cytokine concentrations correlating with outcome in sepsis and trauma. Despite what appears to be a consensus that plasma cytokine concentrations should be useful in the clinical setting, only two cytokines, IL-6 and procalcitonin, have approached routine clinical use. IL-6 has been used as a research tool for entry into sepsis-intervention trials, while procalcitonin is being used clinically at a large number of institutions to distinguish sepsis from other inflammatory processes. For most cytokines, the relative lack of sensitivity and specificity of individual or multiplex cytokine measurements has hindered their utility to predict clinical trajectory in individual patients. The problem rests with a general misunderstanding of cytokine biology, failing to appreciate the general paracrine nature of these mediators, the presence of binding proteins, chaperones and inhibitors in the plasma, and the rapid clearance of these proteins by binding to cell receptors and clearance predominantly by the kidney. The future of using plasma cytokine measurements as an indicator of sepsis/trauma severity or predicting outcome is generally behind us, although there is optimism that procalcitonin measurements may ultimately prove to have utility in the diagnosis of severe sepsis.
Collapse
Affiliation(s)
- Lori F Gentile
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610-0019, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The role of extracorporeal therapies (ECTs) in sepsis is unclear and is a strongly debated topic in critical-care medicine. Unfortunately, much of this debate arises because we lack a clear understanding of what defines the stage and severity of the disease, and the pivotal pathophysiological events dictating outcomes. In the absence of this knowledge, ECTs remain among a large group of therapies with high promise but unproven efficacy.
Collapse
Affiliation(s)
- Ashutosh M Shukla
- Division of Nephrology, University of Arkansas for Medical Sciences, 4301 W. Markham Street Slot 501, Little Rock, Arkansas 72205, USA.
| |
Collapse
|