1
|
Song Z, Li H, Zhang J, Huang Y, Gao S. PLATELET TRAITS AND SEPSIS RISK AND PROGNOSIS: A BIDIRECTIONAL TWO-SAMPLE MENDELIAN RANDOMIZATION STUDY. Shock 2025; 63:520-526. [PMID: 39158958 DOI: 10.1097/shk.0000000000002447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
ABSTRACT Background: Sepsis is a critical medical condition characterized by a dysregulated host response to infection. Platelet abnormalities frequently manifest in sepsis patients, but the causal role of platelets in sepsis remains unclear. This study employed a bidirectional two-sample Mendelian randomization (MR) approach to investigate the causal direction between platelets and sepsis. Methods: MR analysis was used to investigate the causal effect of four platelet traits-platelet count (PLT), platelet crit (PCT), mean platelet volume (MPV), and platelet distribution width (PDW)-on sepsis risk and prognosis. Additionally, the study explored the reverse causality, assessing the impact of sepsis on these platelet traits. Genetic variants from large-scale genome-wide association studies served as instrumental variables to infer causality. Sensitivity analyses and heterogeneity tests were conducted to ensure the validity and robustness of the results. Results: Genetically predicted decreased PCT (OR = 0.938, P = 0.044) and MPV (OR = 0.410, P = 0.006) were associated with an increased risk of sepsis. In the reverse direction, 28-day sepsis mortality was significantly associated with decreased PLT (OR = 0.986, P = 0.034). No significant causal relationships were observed between sepsis and other platelet traits. Conclusions: This study suggests a causal association between low PCT and MPV levels and increased risk of sepsis. Additionally, sepsis with a poor prognosis was causally linked to decreased PLT. These findings provide novel evidence for the causal relationship between platelet traits and sepsis.
Collapse
Affiliation(s)
- Zhonghai Song
- Department of Pre-hospital Emergency, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei, China
| | - Hua Li
- Department of Gastrointestinal Oncology Surgery, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei, China
| | - Jing Zhang
- Department of Neurology, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei, China
| | | | | |
Collapse
|
2
|
Doyle BJ, Kelsey LJ, Shelverton C, Abbate G, Ainola C, Sato N, Livingstone S, Bouquet M, Passmore MR, Wilson ES, Colombo S, Sato K, Liu K, Heinsar S, Wildi K, Carr PJ, Suen J, Fraser J, Li Bassi G, Keogh S. Design, development and preliminary assessment in a porcine model of a novel peripheral intravenous catheter aimed at reducing early failure rates. J Vasc Access 2024; 25:790-799. [PMID: 36281219 DOI: 10.1177/11297298221127760] [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: 02/18/2024] Open
Abstract
BACKGROUND Peripheral intravenous catheters (PIVCs) are the most commonly used invasive medical device, yet despite best efforts by end-users, PIVCs experience unacceptably high early failure rates. We aimed to design a new PIVC that reduces the early failure rate of in-dwelling PIVCs and we conducted preliminary tests to assess its efficacy and safety in a porcine model of intravenous access. METHODS We used computer-aided design and simulation to create a PIVC with a ramped tip geometry, which directs the infused fluid away from the vein wall; we called the design the FloRamp™. We created FloRamp prototypes (test device) and tested them against a market-leading device (BD Insyte™; control device) in a highly-controlled setting with five insertion sites per device in four pigs. We measured resistance to infusion and visual infusion phlebitis (VIP) every 6 h and terminated the experiment at 48 h. Veins were harvested for histology and seven pathological markers were assessed. RESULTS Computer simulations showed that the optimum FloRamp tip reduced maximum endothelial shear stress by 60%, from 12.7 Pa to 5.1 Pa, compared to a typical PIVC tip and improved the infusion dynamics of saline in the blood stream. In the animal study, we found that 2/5 of the control devices were occluded after 24 h, whereas all test devices remained patent and functional. The FloRamp created less resistance to infusion (0.73 ± 0.81 vs 0.47 ± 0.50, p = 0.06) and lower VIP scores (0.60 ± 0.93 vs 0.31 ± 0.70, p = 0.09) than the control device, although neither findings were significantly different. Histopathology revealed that 5/7 of the assessed markers were lower in veins with the FloRamp. CONCLUSIONS Herein we report preliminary assessment of a novel PIVC design, which could be advantageous in clinical settings through decreased device occlusion and reduced early failure rates.
Collapse
Affiliation(s)
- Barry J Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
- School of Engineering, The University of Western Australia, Perth, Western Australia, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, UK
| | - Lachlan J Kelsey
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
- School of Engineering, The University of Western Australia, Perth, Western Australia, Australia
| | | | - Gabriella Abbate
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Noriko Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Samantha Livingstone
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Mahe Bouquet
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Emily S Wilson
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Sebastiano Colombo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Kei Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Keibun Liu
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Silver Heinsar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- St Andrews War Memorial Hospital, Spring Hill, Queensland, Australia
| | - Karin Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Cardiovascular Research Institute Basel, University Hospital of Basel and University Basel, Switzerland
| | - Peter J Carr
- Alliance for Vascular Access Teaching and Research (AVATAR) Group, Menzies Health Institute Queensland, School of Nursing and Midwifery, Griffith University, Brisbane, Queensland, Australia
- School of Nursing and Midwifery, University of Galway, Galway, Ireland
| | - Jacky Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - John Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Queensland University of Technology, Brisbane, Queensland, Australia
- Intensive Care Unit, St Andrews War Memorial Hospital, Spring Hill, Queensland, Australia
- Intensive Care Unit, The Wesley Hospital, Uniting Care Hospitals, Auchenflower, Queensland, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Queensland University of Technology, Brisbane, Queensland, Australia
- Intensive Care Unit, St Andrews War Memorial Hospital, Spring Hill, Queensland, Australia
- Intensive Care Unit, The Wesley Hospital, Uniting Care Hospitals, Auchenflower, Queensland, Australia
| | - Samantha Keogh
- Alliance for Vascular Access Teaching and Research (AVATAR) Group, Menzies Health Institute Queensland, School of Nursing and Midwifery, Griffith University, Brisbane, Queensland, Australia
- School of Nursing and Centre for Healthcare Transformation, Queensland University of Technology, Brisbane, Queensland, Australia
- Nursing and Midwifery Research Centre, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
3
|
Gaudreau LI, Stewart EJ. Vasculature-on-a-chip technologies as platforms for advanced studies of bacterial infections. BIOMICROFLUIDICS 2024; 18:021503. [PMID: 38560344 PMCID: PMC10977040 DOI: 10.1063/5.0179281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
Bacterial infections frequently occur within or near the vascular network as the vascular network connects organ systems and is essential in delivering and removing blood, essential nutrients, and waste products to and from organs. In turn, the vasculature plays a key role in the host immune response to bacterial infections. Technological advancements in microfluidic device design and development have yielded increasingly sophisticated and physiologically relevant models of the vasculature including vasculature-on-a-chip and organ-on-a-chip models. This review aims to highlight advancements in microfluidic device development that have enabled studies of the vascular response to bacteria and bacterial-derived molecules at or near the vascular interface. In the first section of this review, we discuss the use of parallel plate flow chambers and flow cells in studies of bacterial adhesion to the vasculature. We then highlight microfluidic models of the vasculature that have been utilized to study bacteria and bacterial-derived molecules at or near the vascular interface. Next, we review organ-on-a-chip models inclusive of the vasculature and pathogenic bacteria or bacterial-derived molecules that stimulate an inflammatory response within the model system. Finally, we provide recommendations for future research in advancing the understanding of host-bacteria interactions and responses during infections as well as in developing innovative antimicrobials for preventing and treating bacterial infections that capitalize on technological advancements in microfluidic device design and development.
Collapse
Affiliation(s)
- Lily Isabelle Gaudreau
- Chemical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | | |
Collapse
|
4
|
de Hond TAP, Hamelink WJ, de Groot MCH, Hoefer IE, Oosterheert JJ, Haitjema S, Kaasjager KAH. Axial light loss of monocytes as a readily available prognostic biomarker in patients with suspected infection at the emergency department. PLoS One 2022; 17:e0270858. [PMID: 35816504 PMCID: PMC9273078 DOI: 10.1371/journal.pone.0270858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/19/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives To evaluate the prognostic value of the coefficient of variance of axial light loss of monocytes (cv-ALL of monocytes) for adverse clinical outcomes in patients suspected of infection in the emergency department (ED). Methods We performed an observational, retrospective monocenter study including all medical patients ≥18 years admitted to the ED between September 2016 and June 2019 with suspected infection. Adverse clinical outcomes included 30-day mortality and ICU/MCU admission <3 days after presentation. We determined the additional value of monocyte cv-ALL and compared to frequently used clinical prediction scores (SIRS, qSOFA, MEWS). Next, we developed a clinical model with routinely available parameters at the ED, including cv-ALL of monocytes. Results A total of 3526 of patients were included. The OR for cv-ALL of monocytes alone was 2.21 (1.98–2.47) for 30-day mortality and 2.07 (1.86–2.29) for ICU/MCU admission <3 days after ED presentation. When cv-ALL of monocytes was combined with a clinical score, the prognostic accuracy increased significantly for all tested scores (SIRS, qSOFA, MEWS). The maximum AUC for a model with routinely available parameters at the ED was 0.81 to predict 30-day mortality and 0.81 for ICU/MCU admission. Conclusions Cv-ALL of monocytes is a readily available biomarker that is useful as prognostic marker to predict 30-day mortality. Furthermore, it can be used to improve routine prediction of adverse clinical outcomes at the ED. Clinical trial registration Registered in the Dutch Trial Register (NTR) und number 6916.
Collapse
Affiliation(s)
- Titus A. P. de Hond
- Department of Internal Medicine and Acute Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| | - Wout J. Hamelink
- Department of Internal Medicine and Acute Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mark C. H. de Groot
- Central Diagnostic Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Imo E. Hoefer
- Central Diagnostic Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan Jelrik Oosterheert
- Department of Internal Medicine and Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Saskia Haitjema
- Central Diagnostic Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Karin A. H. Kaasjager
- Department of Internal Medicine and Acute Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
5
|
Margraf A, Perretti M. Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes. Cells 2022; 11:cells11111824. [PMID: 35681519 PMCID: PMC9180515 DOI: 10.3390/cells11111824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Inflammation is a life-saving immune reaction occurring in response to invading pathogens. Nonetheless, inflammation can also occur in an uncontrolled, unrestricted manner, leading to chronic disease and organ damage. Mechanisms triggering an inflammatory response, hindering such a response, or leading to its resolution are well-studied but so far insufficiently elucidated with regard to precise therapeutic interventions. Notably, as an immune reaction evolves, requirements and environments for immune cells change, and thus cellular phenotypes adapt and shift, leading to the appearance of distinct cellular subpopulations with new functional features. In this article, we aim to highlight properties of, and overarching regulatory factors involved in, the occurrence of immune cell phenotypes with a special focus on neutrophils, macrophages and platelets. Additionally, we point out implications for both diagnostics and therapeutics in inflammation research.
Collapse
|
6
|
Regulation of TLR4 signaling through the TRAF6/sNASP axis by reversible phosphorylation mediated by CK2 and PP4. Proc Natl Acad Sci U S A 2021; 118:2107044118. [PMID: 34789577 DOI: 10.1073/pnas.2107044118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 02/06/2023] Open
Abstract
Recognition of invading pathogens by Toll-like receptors (TLRs) activates innate immunity through signaling pathways that involved multiple protein kinases and phosphatases. We previously demonstrated that somatic nuclear autoantigenic sperm protein (sNASP) binds to TNF receptor-associated factor 6 (TRAF6) in the resting state. Upon TLR4 activation, a signaling complex consisting of TRAF6, sNASP, interleukin (IL)-1 receptor-associated kinase 4, and casein kinase 2 (CK2) is formed. CK2 then phosphorylates sNASP to release phospho-sNASP (p-sNASP) from TRAF6, initiating downstream signaling pathways. Here, we showed that protein phosphatase 4 (PP4) is the specific sNASP phosphatase that negatively regulates TLR4-induced TRAF6 activation and its downstream signaling pathway. Mechanistically, PP4 is directly recruited by phosphorylated sNASP to dephosphorylate p-sNASP to terminate TRAF6 activation. Ectopic expression of PP4 specifically inhibited sNASP-dependent proinflammatory cytokine production and downstream signaling following bacterial lipopolysaccharide (LPS) treatment, whereas silencing PP4 had the opposite effect. Primary macrophages and mice infected with recombinant adenovirus carrying a gene encoding PP4 (Ad-PP4) showed significant reduction in IL-6 and TNF-α production. Survival of Ad-PP4-infected mice was markedly increased due to a better ability to clear bacteria in a sepsis model. These results indicate that the serine/threonine phosphatase PP4 functions as a negative regulator of innate immunity by regulating the binding of sNASP to TRAF6.
Collapse
|
7
|
Rahman M, Ding Z, Rönnow CF, Thorlacius H. Transcriptomic Analysis Reveals Differential Expression of Genes between Lung Capillary and Post Capillary Venules in Abdominal Sepsis. Int J Mol Sci 2021; 22:ijms221910181. [PMID: 34638535 PMCID: PMC8507973 DOI: 10.3390/ijms221910181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022] Open
Abstract
Lung endothelial cell dysfunction plays a central role in septic-induced lung injury. We hypothesized that endothelial cell subsets, capillary endothelial cells (capEC) and post capillary venules (PCV), might play different roles in regulating important pathophysiology in sepsis. In order to reveal global transcriptomic changes in endothelial cell subsets during sepsis, we induced sepsis in C57BL/6 mice by cecal ligation and puncture (CLP). We confirmed that CLP induced systemic and lung inflammation in our model. Endothelial cells (ECs) from lung capillary and PCV were isolated by cell sorting and transcriptomic changes were analyzed by bioinformatic tools. Our analysis revealed that lung capEC are transcriptionally different than PCV. Comparison of top differentially expressed genes (DEGs) of capEC and PCV revealed that capEC responses are different than PCV during sepsis. It was found that capEC are more enriched with genes related to regulation of coagulation, vascular permeability, wound healing and lipid metabolic processes after sepsis. In contrast, PCV are more enriched with genes related to chemotaxis, cell–cell adhesion by integrins, chemokine biosynthesis, regulation of actin filament process and neutrophil homeostasis after sepsis. In addition, we predicted some transcription factor targets that regulate a significant number of DEGs in sepsis. We proposed that targeting certain DEGs or transcriptional factors would be useful in protecting against sepsis-induced lung damage.
Collapse
|
8
|
Dickson K, Malitan H, Lehmann C. Imaging of the Intestinal Microcirculation during Acute and Chronic Inflammation. BIOLOGY 2020; 9:E418. [PMID: 33255906 PMCID: PMC7760140 DOI: 10.3390/biology9120418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Because of its unique microvascular anatomy, the intestine is particularly vulnerable to microcirculatory disturbances. During inflammation, pathological changes in blood flow, vessel integrity and capillary density result in impaired tissue oxygenation. In severe cases, these changes can progress to multiorgan failure and possibly death. Microcirculation may be evaluated in superficial tissues in patients using video microscopy devices, but these techniques do not allow the assessment of intestinal microcirculation. The gold standard for the experimental evaluation of intestinal microcirculation is intravital microscopy, a technique that allows for the in vivo examination of many pathophysiological processes including leukocyte-endothelial interactions and capillary blood flow. This review provides an overview of changes in the intestinal microcirculation in various acute and chronic inflammatory conditions. Acute conditions discussed include local infections, severe acute pancreatitis, necrotizing enterocolitis and sepsis. Inflammatory bowel disease and irritable bowel syndrome are included as examples of chronic conditions of the intestine.
Collapse
Affiliation(s)
- Kayle Dickson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Hajer Malitan
- Department of Anesthesia, Pain and Perioperative Management, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Christian Lehmann
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Anesthesia, Pain and Perioperative Management, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
9
|
Fine N, Gracey E, Dimitriou I, La Rose J, Glogauer M, Rottapel R. GEF-H1 Is Required for Colchicine Inhibition of Neutrophil Rolling and Recruitment in Mouse Models of Gout. THE JOURNAL OF IMMUNOLOGY 2020; 205:3300-3310. [PMID: 33199537 DOI: 10.4049/jimmunol.1900783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/20/2020] [Indexed: 11/19/2022]
Abstract
Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1β production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.
Collapse
Affiliation(s)
- Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Eric Gracey
- Vlaams Institute for Biotechnology Centre for Inflammation Research, 9052 Ghent, Belgium.,Department of Internal Medicine and Pediatrics, University of Ghent, 9000 Ghent, Belgium
| | - Ioannis Dimitriou
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - José La Rose
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Robert Rottapel
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada; .,Department of Medicine, Ontario Institute for Cancer Research, University of Toronto, Toronto, Ontario M5G 1L7, Canada; and.,Division of Rheumatology, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
10
|
A Novel Implementation of Magnetic Levitation to Quantify Leukocyte Size, Morphology, and Magnetic Properties to Identify Patients With Sepsis. Shock 2020; 51:147-152. [PMID: 29561389 DOI: 10.1097/shk.0000000000001139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND We have developed a novel, easily implementable methodology using magnetic levitation to quantify circulating leukocyte size, morphology, and magnetic properties, which may help in rapid, bedside screening for sepsis. OBJECTIVE Our objectives were to describe our methodological approach to leukocyte assessment, and to perform a pilot investigation to test the ability of magnetic levitation to identify and quantify changes in leukocyte size, shape, density, and/or paramagnetic properties in healthy controls and septic patients. METHODS This prospective, observational cohort study was performed in a 56,000/y visit emergency department (ED) and affiliated outpatient phlebotomy laboratory. Inclusion criteria were admittance to the hospital with suspected or confirmed infection for the septic group, and we enrolled the controls from ED/outpatient patients without infection or acute illness. The bench-top experiments were performed using magnetic levitation to visualize the leukocytes. We primary sought to compare septic patients with noninfected controls and secondary to assess the association with sepsis severity. Our covariates were area, length, width, roundness, and standard deviation (SD) of levitation height. We used unpaired t test and area under the curve (AUC) for the assessment of accuracy in distinguishing between septic and control patients. RESULTS We enrolled 39 noninfected controls and 22 septic patients. Our analyses of septic patients compared with controls showed: mean cell area in pixels (px) 562 ± 111 vs. 410 ± 45, P < 0.0001, AUC = 0.89 (0.80-0.98); length (px), 29 ± 2.5 vs. 25 ± 1.9, P < 0.0001, AUC = 0.90 (0.83-0.98); and width (px), 27 ± 2.4 vs. 23 ± 1.5, P < 0.0001, AUC = 0.92 (0.84-0.99). Cell roundness: 2.1 ± 1.0 vs. 2.2 ± 1.2, P = 0.8, AUC = 0.51. SD of the levitation height (px) was 72 ± 25 vs. 47 ± 16, P < 0.001, AUC = 0.80 (0.67-0.93). CONCLUSIONS Septic patients had circulating leukocytes with especially increased size parameters, which distinguished sepsis from noninfected patients with promising high accuracy. This portal-device compatible technology shows promise as a potential bedside diagnostic.
Collapse
|
11
|
Kawamoto E, Nago N, Okamoto T, Gaowa A, Masui-Ito A, Sakakura Y, Akama Y, Soe ZY, Prajuabjinda O, Darkwah S, Appiah MG, Myint PK, Obeng G, Park EJ, Imai H, Shimaoka M. Anti-adhesive effects of human soluble thrombomodulin and its domains. Biochem Biophys Res Commun 2019; 511:312-317. [PMID: 30777333 DOI: 10.1016/j.bbrc.2019.02.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 02/08/2019] [Indexed: 11/29/2022]
Abstract
We reported previously that leukocyte β2 integrins (LFA-1 and Mac-1) bind to the serine/threonine-rich domain of thrombomodulin (TM) expressed on vascular endothelial cells (VECs). Recombinant human soluble TM (rhsTM, TMD123) has been approved as a therapeutic drug for septic disseminated intravascular coagulation. However, the roles of TMD123 on the adhesion of leukocyte integrins to VECs remain unclear. In the current study, we have revealed that an integrin-dependent binding between human peripheral blood mononuclear cells (PBMCs) and VECs was inhibited by TMD123. Next, using mutant proteins composed of isolated TM extracellular domains, we examined the structural characteristics responsible for the anti-adhesion properties of TMD123. Namely, we investigated whether the effects of the binding of TM and leukocytes was inhibited by the administration of TMD123. In fact, we confirmed that TMD123, TMD1, and TMD3 inhibited the binding of PBMCs to the immobilized recombinant proteins TMD123 and TMD3. These results indicate that TMD123 inhibited the adhesion of leukocytes to endothelial cells via β2 integrins and endothelial TM. Moreover, since TMD1 might bind to leukocytes via other adhesion receptors than integrins, TMD1 and TMD3 appear to inhibit leukocyte binding to TM on VECs via different mechanisms. In summary, TMD123 (rhsTM), TMD1 or TMD3 is a promising treatment option for sepsis that attenuates integrin-dependent binding of leukocytes to VECs, and may inhibit the undesirable adhesion and migration of leukocytes to VECs in sepsis.
Collapse
Affiliation(s)
- Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan.
| | - Nodoka Nago
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan; Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science, 1001-1 Kishioka, Suzuka-city, Mie, 510-0293, Japan
| | - Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane, 693-8501, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Asami Masui-Ito
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Yosuke Sakakura
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Yuichi Akama
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Zay Yar Soe
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Onmanee Prajuabjinda
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Samuel Darkwah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Michael G Appiah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Phyoe Kyawe Myint
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Gideon Obeng
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Hiroshi Imai
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie, 514-8507, Japan
| |
Collapse
|
12
|
Hawthorn A, Bulmer AC, Mosawy S, Keogh S. Implications for maintaining vascular access device patency and performance: Application of science to practice. J Vasc Access 2019; 20:461-470. [DOI: 10.1177/1129729818820200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction:Vascular access devices are commonly inserted devices that facilitate the administration of fluids and drugs, as well as blood sampling. Despite their common use in clinical settings, these devices are prone to occlusion and failure, requiring replacement and exposing the patient to ongoing discomfort/pain, local vessel inflammation and risk of infection. A range of insertion and maintenance strategies are employed to optimize device performance; however, the evidence base for many of these mechanisms is limited and the mechanisms contributing to the failure of these devices are largely unknown.Aims/objectives:(1) To revisit existing understanding of blood, vessel physiology and biological fluid dynamics; (2) develop an understanding of the implications that different clinical practices have on vessel health, and (3) apply these understandings to vascular access device research and practice.Method:Narrative review of biomedical and bioengineering studies related to vascular access practice.Results/outcomes:Current vascular access device insertion and maintenance practice and policy are variable with limited clinical evidence to support the theoretical assumptions underpinning these regimens. This review demonstrates the physiological response to vascular access device insertion, flushing and infusion on the vein, blood components and blood flow. These appear to be associated with changes in intravascular fluid dynamics. Variable forces are at play that impact blood componentry and the endothelium. These may explain the mechanisms contributing to vascular access failure.Conclusion:This review provides an update to our current knowledge and understanding of vascular physiology and the hemodynamic response, challenging some previously held assumptions regarding vascular access device maintenance, which require further investigation.
Collapse
Affiliation(s)
- Alexandra Hawthorn
- School of Nursing, Queensland University of Technology, Brisbane, QLD, Australia
- School of Medicine, Griffith University, Brisbane, QLD, Australia
- Alliance for Vascular Access Teaching and Research (AVATAR), Menzies Health Institute Queensland, Griffith University, Brisbane, QLD, Australia
| | - Andrew C Bulmer
- School of Medicine, Griffith University, Brisbane, QLD, Australia
- Alliance for Vascular Access Teaching and Research (AVATAR), Menzies Health Institute Queensland, Griffith University, Brisbane, QLD, Australia
| | - Sapha Mosawy
- School of Medicine, Griffith University, Brisbane, QLD, Australia
| | - Samantha Keogh
- School of Nursing, Queensland University of Technology, Brisbane, QLD, Australia
- Alliance for Vascular Access Teaching and Research (AVATAR), Menzies Health Institute Queensland, Griffith University, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Lin B, Jia X, Xie Z, Su T, Wei Y, Tang J, Yang C, Cui C, Liu J. Vascular Endothelial Cells Activate Peripheral Natural Killer T Cells and Participate in Regulation of Downstream Immune Cascades in Patients with Sepsis. Med Sci Monit 2018; 24:7387-7398. [PMID: 30324936 PMCID: PMC6199819 DOI: 10.12659/msm.911466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background This study investigated the effect of supernatant of endothelial cells stimulated by peripheral blood serum from sepsis patients on phenotype and function of peripheral NKT cells. Material/Methods Twenty-one patients with sepsis and 21 healthy subjects were included. Peripheral blood (5 ml) was collected from all patients and healthy subjects. To isolate peripheral blood mononuclear cells (PBMCs), Ficoll lymphocyte separation solution was used. Flow cytometry was carried out to determine NKT cell ratio, activity, and cytokine secretion. Human umbilical vein endothelial cells were cultured with serum from sepsis patients for 48 h before changing to fresh medium, and supernatant was collected. The supernatant was used to co-culture PBMCs before analyzing NKT activity and cytokines. Results The ratios of CD3-CD56+NK cells and CD3+CD56+NKT cells were increased in peripheral blood from sepsis patients. Surface receptors p30, G2D, and p44 of CD3+CD56+NKT cells were elevated, while inhibitory receptors NKG2A and 158b were decreased. CD4+ NKT cells in peripheral blood from sepsis patients were enhanced. GranB, IFN-γ, IL-4, and IL-17 in NKT cells from sepsis patients were up-regulated. After co-culture with vascular endothelial cells treated with sepsis serum, expression of p30 and G2D in NKT cells was upregulated, and number of TCRVα24-positive cells was increased. In addition, ratio of CD4+NKT cells was increased, and intracellular expression of IL-4 and IFN-γ was elevated. Conclusions The study demonstrates that the level of NKT cells in peripheral blood from sepsis patients is increased, and their activity is enhanced. In addition, vascular endothelial cells from sepsis patients can regulate the activity of NKT cells.
Collapse
Affiliation(s)
- Bing Lin
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Xinju Jia
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Zuohua Xie
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ting Su
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ying Wei
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jiping Tang
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Chengzhi Yang
- Graduate School, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Chuanbao Cui
- Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jinxiang Liu
- Department of Intensive Medicine, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
14
|
Leligdowicz A, Chun LF, Jauregui A, Vessel K, Liu KD, Calfee CS, Matthay MA. Human pulmonary endothelial cell permeability after exposure to LPS-stimulated leukocyte supernatants derived from patients with early sepsis. Am J Physiol Lung Cell Mol Physiol 2018; 315:L638-L644. [PMID: 30024307 DOI: 10.1152/ajplung.00286.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic immune activation is the hallmark of sepsis, which can result in endothelial injury and the acute respiratory distress syndrome (ARDS). The aim of this study was to investigate heterogeneity in sepsis-mediated endothelial permeability using primary human pulmonary microvascular endothelial cells (HPMECs) and the electric cell-substrate impedance sensing (ECIS) platform. After plasma removal, cellular component of whole blood from 35 intensive care unit (ICU) patients with early sepsis was diluted with media and stimulated with either lipopolysaccharide (LPS) or control media. Resulting supernatants were cocultured with HPMECs seeded on ECIS plates, and resistance was continually measured. A decrease in resistance signified increased permeability. After incubation, HPMECs were detached and cell adhesion proteins were quantified using flow cytometry and immunohistochemistry, and gene expression was analyzed with quantitative PCR. Significant heterogeneity in endothelial permeability after exposure to supernatants of LPS-stimulated leukocytes was identified. ICU patients with sepsis stratified into one of the following three groups: minimal (9/35, 26%), intermediate (18/35, 51%), and maximal (8/35, 23%) permeability. Maximal permeability was associated with increased intercellular adhesion molecule-1 protein and mRNA expression and decreased vascular endothelial-cadherin mRNA expression. These findings indicate that substantial heterogeneity in pulmonary endothelial permeability is induced by supernatants of LPS-stimulated leukocytes derived from patients with early sepsis and provide insights into some of the mechanisms that induce lung vascular injury. In addition, this in vitro model of lung endothelial permeability from LPS-stimulated leukocytes may be a useful method for testing therapeutic agents that could mitigate endothelial injury in early sepsis.
Collapse
Affiliation(s)
- Aleksandra Leligdowicz
- Cardiovascular Research Institute, University of California , San Francisco, California.,Interdepartmental Division of Critical Care Medicine, University of Toronto , Toronto, Ontario , Canada
| | - Lauren F Chun
- Cardiovascular Research Institute, University of California , San Francisco, California
| | - Alejandra Jauregui
- Cardiovascular Research Institute, University of California , San Francisco, California
| | - Kathryn Vessel
- Cardiovascular Research Institute, University of California , San Francisco, California
| | - Kathleen D Liu
- Cardiovascular Research Institute, University of California , San Francisco, California.,Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California , San Francisco, California
| | - Carolyn S Calfee
- Cardiovascular Research Institute, University of California , San Francisco, California.,Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California , San Francisco, California
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California , San Francisco, California.,Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California , San Francisco, California.,Departments of Medicine and Anesthesia, University of California , San Francisco, California
| |
Collapse
|
15
|
Finsterbusch M, Schrottmaier WC, Kral-Pointner JB, Salzmann M, Assinger A. Measuring and interpreting platelet-leukocyte aggregates. Platelets 2018; 29:677-685. [PMID: 29461910 PMCID: PMC6178087 DOI: 10.1080/09537104.2018.1430358] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets, besides their specialised role in haemostasis and atherothrombosis, actively modulate innate and adaptive immune responses with crucial roles in immune surveillance, inflammation and host defence during infection. An important prerequisite for platelet-mediated changes of immune functions involves direct engagement with different types of leukocytes. Indeed, increased platelet-leukocyte aggregates (PLAs) within the circulation and/or locally at the site of inflammation represent markers of many thrombo-inflammatory diseases, such as cardiovascular diseases, acute lung injury, renal and cerebral inflammation. Therefore, measurement of PLAs could provide an attractive and easily accessible prognostic and/or diagnostic tool for many diseases. To measure PLAs in different (patho-)physiological settings in human and animal models flow cytometric and microscopic approaches have been applied. These techniques represent complementary tools to study different aspects relating to the involvement of leukocyte subtypes and molecules, as well as location of PLAs within tissues, dynamics of their interactions and/or dynamic changes in leukocyte and platelet behaviour. This review summarises various approaches to measure and interpret PLAs and discusses potential experimental factors influencing platelet binding to leukocytes. Furthermore, we summarise insights gained from studies regarding the underlying mechanism of platelet-leukocyte interactions and discuss implications of these interactions in health and disease.
Collapse
Affiliation(s)
- Michaela Finsterbusch
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Waltraud C Schrottmaier
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Julia B Kral-Pointner
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Manuel Salzmann
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Alice Assinger
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
16
|
Choi W, Kim HM, Park S, Yeom E, Doh J, Lee SJ. Variation in wall shear stress in channel networks of zebrafish models. J R Soc Interface 2017; 14:rsif.2016.0900. [PMID: 28148768 DOI: 10.1098/rsif.2016.0900] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023] Open
Abstract
Physiological functions of vascular endothelial cells (ECs) vary depending on wall shear stress (WSS) magnitude, and the functional change affects the pathologies of various cardiovascular systems. Several in vitro and in vivo models have been used to investigate the functions of ECs under different WSS conditions. However, these models have technical limitations in precisely mimicking the physiological environments of ECs and monitoring temporal variations of ECs in detail. Although zebrafish (Danio rerio) has several strategies to overcome these technical limitations, zebrafish cannot be used as a perfect animal model because applying various WSS conditions on blood vessels of zebrafish is difficult. This study proposes a new zebrafish model in which various WSS can be applied to the caudal vein. The WSS magnitude is controlled by blocking some parts of blood-vessel networks. The accuracy and reproducibility of the proposed method are validated using an equivalent circuit model of blood vessels in zebrafish. The proposed method is applied to lipopolysaccharide (LPS)-stimulated zebrafish as a typical application. The proposed zebrafish model can be used as an in vivo animal model to investigate the relationship between WSS and EC physiology or WSS-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Woorak Choi
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| | - Hye Mi Kim
- Division of Integrative Biosciences and Biotechnology (IBB), Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Sungho Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| | - Eunseop Yeom
- School of Mechanical Engineering, Pusan National University, Busan, South Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| | - Sang Joon Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| |
Collapse
|
17
|
Mechanisms of endothelial activation in sepsis and cell culture models to study the heterogeneous host response. Int J Artif Organs 2017; 40:9-14. [PMID: 28218355 DOI: 10.5301/ijao.5000560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2017] [Indexed: 01/26/2023]
Abstract
Sepsis is currently viewed as a fundamental disintegration of control functions from intracellular signalling to immunoregulatory and neuroendocrine mechanisms. The immediate threat in sepsis is invasive infection, and the need to activate immune defense mechanisms to clear the pathogen before irreparable damage occurs. In the process of pathogen elimination, however, the systemic host response to infection may cause collateral damage to the endothelium and may lead to the destruction of host tissues.A number of experimental models have been developed to monitor endothelial activation and to study endothelial dysfunction under septic conditions. Here, we review the application of these models to assess the highly variable host response in sepsis and to investigate the efficacy of adsorbent-based extracorporeal therapies. We also highlight the need for efficient diagnostic tools, which are indispensable to select patients who are likely to benefit from distinct adjunctive therapies.
Collapse
|
18
|
Wu J, Li X, Huang L, Jiang S, Tu F, Zhang X, Ma H, Li R, Li C, Li Y, Ding Z, Liu L. HSPA12B inhibits lipopolysaccharide-induced inflammatory response in human umbilical vein endothelial cells. J Cell Mol Med 2014; 19:544-54. [PMID: 25545050 PMCID: PMC4369812 DOI: 10.1111/jcmm.12464] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 09/09/2014] [Indexed: 01/23/2023] Open
Abstract
Heat shock protein A12B (HSPA12B) is a newly discovered member of the HSP70 protein family. This study investigated the effects of HSPA12B on lipopolysaccharide (LPS)-induced inflammatory responses in human umbilical vein endothelial cells (HUVECs) and the possible mechanisms involved. A HUVECs inflammatory model was induced by LPS. Overexpression of HSPA12B in HUVECs was achieved by infection with recombinant adenoviruses encoding green fluorescence protein-HSPA12B. Knockdown of HSPA12B was achieved by siRNA technique. Twenty four hours after virus infection or siRNA transfection, HUVECs were stimulated with 1 μg/ml LPS for 4 hrs. Endothelial cell permeability ability was determined by transwell permeability assay. The binding rate of human neutrophilic polymorphonuclear leucocytes (PMN) with HUVECs was examined using myeloperoxidase assay. Cell migrating ability was determined by the wound-healing assay. The mRNA and protein expression levels of interested genes were analyzed by RT-qPCR and Western blot, respectively. The release of cytokines interleukin-6 and tumour necrosis factor-α was measured by ELISA. HSPA12B suppressed LPS-induced HUVEC permeability and reduced PMN adhesion to HUVECs. HSPA12B also inhibited LPS-induced up-regulation of adhesion molecules and inflammatory cytokine expression. By contrast, knockdown of HSPA12B enhanced LPS-induced increases in the expression of adhesion molecules and inflammatory cytokines. Moreover, HSPA12B activated PI3K/Akt signalling pathway and pharmacological inhibition of this pathway by Wortmannin completely abrogated the protection of HSPA12B against inflammatory response in HUVECs. Our results suggest that HSPA12B attenuates LPS-induced inflammatory responses in HUVECs via activation of PI3K/Akt signalling pathway.
Collapse
Affiliation(s)
- Jun Wu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Harris DG, Quinn KJ, Dahi S, Burdorf L, Azimzadeh AM, Pierson RN. Lung xenotransplantation: recent progress and current status. Xenotransplantation 2014; 21:496-506. [PMID: 25040467 DOI: 10.1111/xen.12116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Xenotransplantation has undergone important progress in controlling initial hyperacute rejection in many preclinical models, with some cell, tissue, and organ xenografts advancing toward clinical trials. However, acute injury, driven primarily by innate immune and inflammatory responses, continues to limit results in lung xenograft models. The purpose of this article is to review the current status of lung xenotransplantation--including the seemingly unique challenges posed by this organ-and summarize proven and emerging means of overcoming acute lung xenograft injury.
Collapse
Affiliation(s)
- Donald G Harris
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|
20
|
de Stoppelaar SF, van 't Veer C, van der Poll T. The role of platelets in sepsis. Thromb Haemost 2014; 112:666-77. [PMID: 24966015 DOI: 10.1160/th14-02-0126] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/16/2014] [Indexed: 01/01/2023]
Abstract
Platelets are small circulating anucleate cells that are of crucial importance in haemostasis. Over the last decade, it has become increasingly clear that platelets play an important role in inflammation and can influence both innate and adaptive immunity. Sepsis is a potentially lethal condition caused by detrimental host response to an invading pathogen. Dysbalanced immune response and activation of the coagulation system during sepsis are fundamental events leading to sepsis complications and organ failure. Platelets, being major effector cells in both haemostasis and inflammation, are involved in sepsis pathogenesis and contribute to sepsis complications. Platelets catalyse the development of hyperinflammation, disseminated intravascular coagulation and microthrombosis, and subsequently contribute to multiple organ failure. Inappropriate accumulation and activity of platelets are key events in the development of sepsis-related complications such as acute lung injury and acute kidney injury. Platelet activation readouts could serve as biomarkers for early sepsis recognition; inhibition of platelets in septic patients seems like an important target for immune-modulating therapy and appears promising based on animal models and retrospective human studies.
Collapse
Affiliation(s)
- Sacha F de Stoppelaar
- Sacha F. de Stoppelaar, MD, Academic Medical Centre, Centre of Experimental and Molecular Medicine, Meibergdreef 9, Room G2-130, 1105 AZ Amsterdam, the Netherlands, Tel.: +31 20 5665910, Fax: +31 20 6977192, E-mail:
| | | | | |
Collapse
|
21
|
Smith AM, Prabhakarpandian B, Pant K. Generation of shear adhesion map using SynVivo synthetic microvascular networks. J Vis Exp 2014. [PMID: 24893648 DOI: 10.3791/51025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Cell/particle adhesion assays are critical to understanding the biochemical interactions involved in disease pathophysiology and have important applications in the quest for the development of novel therapeutics. Assays using static conditions fail to capture the dependence of adhesion on shear, limiting their correlation with in vivo environment. Parallel plate flow chambers that quantify adhesion under physiological fluid flow need multiple experiments for the generation of a shear adhesion map. In addition, they do not represent the in vivo scale and morphology and require large volumes (~ml) of reagents for experiments. In this study, we demonstrate the generation of shear adhesion map from a single experiment using a microvascular network based microfluidic device, SynVivo-SMN. This device recreates the complex in vivo vasculature including geometric scale, morphological elements, flow features and cellular interactions in an in vitro format, thereby providing a biologically realistic environment for basic and applied research in cellular behavior, drug delivery, and drug discovery. The assay was demonstrated by studying the interaction of the 2 µm biotin-coated particles with avidin-coated surfaces of the microchip. The entire range of shear observed in the microvasculature is obtained in a single assay enabling adhesion vs. shear map for the particles under physiological conditions.
Collapse
Affiliation(s)
| | | | - Kapil Pant
- Biomedical Technology, CFD Research Corporation
| |
Collapse
|
22
|
Lewis SM, Khan N, Beale R, Treacher DF, Brown KA. Depletion of blood neutrophils from patients with sepsis: treatment for the future? Int Immunopharmacol 2013; 17:1226-32. [PMID: 24144812 DOI: 10.1016/j.intimp.2013.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Organ failure arising from severe sepsis accounts for nearly 6 million deaths worldwide per annum. At present there are no specific pharmacological agents available for its treatment and identifying a suitable therapeutic target is urgently needed. Neutrophils appear to be contributing directly to pulmonary damage in severe forms of lung injury and indirectly to the failure of other organs. Blood neutrophils from patients with sepsis possess a phenotype that is indicative of activation and our results show that neutrophils isolated from patients with sepsis exhibit a supranormal adherence to endothelial monolayers treated with pro-inflammatory cytokines. Additional studies reveal that the patients' cells are highly efficient at releasing IL-8. We also demonstrate that organ function is improved upon removing neutrophils from the circulation. In this article we propose that in severe sepsis there is a subpopulation of neutrophils which is actively engaged in pathological insult. The phenotypic characterisation of this subset may provide a novel therapeutic strategy for sepsis that could lead to patient benefit.
Collapse
Affiliation(s)
- Sion M Lewis
- Intensive Care Unit, Guy's and St. Thomas' NHS Foundation Trust, London, UK; Vascular Immunology, King's College London, UK
| | | | | | | | | |
Collapse
|
23
|
Fox ED, Heffernan DS, Cioffi WG, Reichner JS. Neutrophils from critically ill septic patients mediate profound loss of endothelial barrier integrity. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R226. [PMID: 24099563 PMCID: PMC4057230 DOI: 10.1186/cc13049] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 09/06/2013] [Indexed: 12/30/2022]
Abstract
Introduction Sepsis is characterized by systemic immune activation and neutrophil-mediated endothelial barrier integrity compromise, contributing to end-organ dysfunction. Studies evaluating endothelial barrier dysfunction induced by neutrophils from septic patients are lacking, despite its clinical significance. We hypothesized that septic neutrophils would cause characteristic patterns of endothelial barrier dysfunction, distinct from experimental stimulation of normal neutrophils, and that treatment with the immunomodulatory drug β-glucan would attenuate this effect. Methods Blood was obtained from critically ill septic patients. Patients were either general surgery patients (Primary Sepsis (PS)) or those with sepsis following trauma (Secondary Sepsis (SS)). Those with acute respiratory distress syndrome (ARDS) were identified. Healthy volunteers served as controls. Neutrophils were purified and aliquots were untreated, or treated with fMLP or β-glucan. Endothelial cells were grown to confluence and activated with tissue necrosis factor (TNF)-α . Electric Cell-substrate Impedance Sensing (ECIS) was used to determine monolayer resistance after neutrophils were added. Groups were analyzed by two-way analysis of variance (ANOVA). Results Neutrophils from all septic patients, as well as fMLP-normal neutrophils, reduced endothelial barrier integrity to a greater extent than untreated normal neutrophils (normalized resistance of cells from septic patients at 30 mins = 0.90 ± 0.04; at 60 mins = 0.73 ± 0.6 and at 180 mins = 0.56 ± 0.05; p < 0.05 vs normal). Compared to untreated PS neutrophils, fMLP-treated PS neutrophils caused further loss of barrier function at all time points; no additive effect was noted in stimulation of SS neutrophils beyond 30 min. Neutrophils from ARDS patients caused greater loss of barrier integrity than those from non-ARDS patients, despite similarities in age, sex, septic source, and neutrophil count. Neutrophils obtained after resolution of sepsis caused less barrier dysfunction at all time points. β-glucan treatment of septic patients’ neutrophils attenuated barrier compromise, rendering the effect similar to that induced by neutrophils obtained once sepsis had resolved. Conclusions Neutrophils from septic patients exert dramatic compromise of endothelial barrier integrity. This pattern is mimicked by experimental activation of healthy neutrophils. The effect of septic neutrophils on the endothelium depends upon the initial inflammatory event, correlates with organ dysfunction and resolution of sepsis, and is ameliorated by β-glucan.
Collapse
|
24
|
Rimmelé T, Kaynar AM, McLaughlin JN, Bishop JV, Fedorchak MV, Chuasuwan A, Peng Z, Singbartl K, Frederick DR, Zhu L, Carter M, Federspiel WJ, Zeevi A, Kellum JA. Leukocyte capture and modulation of cell-mediated immunity during human sepsis: an ex vivo study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R59. [PMID: 23531333 PMCID: PMC3672497 DOI: 10.1186/cc12587] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 03/18/2013] [Indexed: 01/06/2023]
|
25
|
Ploppa A, Kampmann M, Johannes T, Haeberle HA, Nohé B. Effects of different leukocyte subpopulations and flow conditions on leukocyte accumulation during reperfusion. J Vasc Res 2012; 49:169-80. [PMID: 22398918 DOI: 10.1159/000335147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 11/11/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The study examined the interdependent effects of shear stress and different leukocyte subpopulations on endothelial cell activation and cell interactions during low flow and reperfusion. METHODS Human umbilical venous endothelial cells were perfused with either neutrophils or monocytes at different shear stress (2-0.25 dyn/cm(2)) and adhesion was quantified by microscopy. Effects of adherent neutrophils and monocytes on endothelial cell adhesion molecule expression were analyzed by flow cytometry after 4-hour static coincubation. After coincubation, the cocultures were reperfused with labeled neutrophils at 2 dyn/cm(2) and their adhesion was quantified selectively. For the control, endothelium monocultures with and without lipopolysaccharide activation were used. RESULTS At 2 dyn/cm(2), adhesion did not exceed baseline levels on nonactivated endothelium. Decreasing shear stress to 0.25 dyn/cm(2) largely increased the adhesion of both leukocyte subpopulations, similar to the effect of lipopolysaccharide at 2 dyn/cm(2). However, only adherent monocytes increased adhesion molecule expression, whereas neutrophils had no effect. As a functional consequence, adherent monocytes largely increased neutrophil adhesion during reperfusion, whereas adherent neutrophils did not. CONCLUSION Compromised shear stress is an autonomous trigger of leukocyte adhesion even in the absence of additional activators. Exceeding this immediate effect, adherent monocytes induce further endothelial activation and enhance further neutrophil adhesion during reperfusion.
Collapse
Affiliation(s)
- Annette Ploppa
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Eberhard-Karls University, Tübingen, Germany
| | | | | | | | | |
Collapse
|
26
|
Kandasamy K, Sahu G, Parthasarathi K. Real-time imaging reveals endothelium-mediated leukocyte retention in LPS-treated lung microvessels. Microvasc Res 2012; 83:323-31. [PMID: 22342350 DOI: 10.1016/j.mvr.2012.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 12/31/2011] [Accepted: 01/31/2012] [Indexed: 01/01/2023]
Abstract
Endotoxemia, a major feature of sepsis, is a common cause of acute lung injury and initiates rapid accumulation of leukocytes in the lung vasculature. Endothelial mechanisms that underlie this accumulation remain unclear, as current experimental models of endotoxemia are less suitable for targeted activation of the endothelium. Toward elucidating this, we used the isolated blood-perfused rat lung preparation. With a microcatheter inserted through a left atrial cannula, we cleared blood cells from a small lung region and then infused lipopolysaccharide (LPS) into microvessels. After a Ringer's wash to remove residual LPS, we infused fluorescently-labeled autologous leukocytes and imaged their transit through the treated microvessels. Image analysis revealed that leukocytes infused 90 min after LPS treatment were retained more in treated venules and capillaries than untreated vessels. Further, pretreatment with either the intercellular adhesion molecule-1 (ICAM-1) mAb or polymyxin-B blunted LPS-induced leukocyte retention in both microvessel segments. In addition, retention of leukocytes treated ex vivo with LPS in LPS-treated microvessels was higher compared to retention of untreated leukocytes. In situ immunofluorescence experiments revealed that LPS significantly increased microvessel ICAM-1 expression at 90 min post treatment. Polymyxin pretreatment inhibited this increase. Taken together, the data suggest that LPS increased leukocyte retention in both venules and capillaries and this response was mediated by the increased expression of endothelial ICAM-1. Thus, endothelial mechanisms may themselves play a major role in LPS-induced leukocyte retention in lung microvessels. Blunting the endothelial responses may mitigate endotoxin-induced morbidity.
Collapse
Affiliation(s)
- Kathirvel Kandasamy
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | |
Collapse
|
27
|
Yan T, Li Q, Zhou H, Zhao Y, Yu S, Xu G, Yin Z, Li Z, Zhao Z. Gu-4 suppresses affinity and avidity modulation of CD11b and improves the outcome of mice with endotoxemia and sepsis. PLoS One 2012; 7:e30110. [PMID: 22319560 PMCID: PMC3271073 DOI: 10.1371/journal.pone.0030110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/09/2011] [Indexed: 02/06/2023] Open
Abstract
Background Systemic leukocyte activation and disseminated leukocyte adhesion will impair the microcirculation and cause severe decrements in tissue perfusion and organ function in the process of severe sepsis. Gu-4, a lactosyl derivative, could selectively target CD11b to exert therapeutic effect in a rat model of severe burn shock. Here, we addressed whether Gu-4 could render protective effects on septic animals. Methodology/Principal Findings On a murine model of endotoxemia induced by lipopolysaccharide (LPS), we found that the median effective dose (ED50) of Gu-4 was 0.929 mg/kg. In vivo treatment of Gu-4 after LPS challenge prominently attenuated LPS-induced lung injury and decreased lactic acid level in lung tissue. Using the ED50 of Gu-4, we also demonstrated that Gu-4 treatment significantly improved the survival rate of animals underwent sepsis induced by cecal ligation and puncture. By adhesion and transwell migration assays, we found that Gu-4 treatment inhibited the adhesion and transendothelial migration of LPS-stimulated THP-1 cells. By flow cytometry and microscopy, we demonstrated that Gu-4 treatment inhibited the exposure of active I-domain and the cluster formation of CD11b on the LPS-stimulated polymorphonuclear leukocytes. Western blot analyses further revealed that Gu-4 treatment markedly inhibited the activation of spleen tyrosine kinase in LPS-stimulated THP-1 cells. Conclusions/Significance Gu-4 improves the survival of mice underwent endotoxemia and sepsis, our in vitro investigations indicate that the possible underlying mechanism might involve the modulations of the affinity and avidity of CD11b on the leukocyte. Our findings shed light on the potential use of Gu-4, an interacting compound to CD11b, in the treatment of sepsis and septic shock.
Collapse
Affiliation(s)
- TingTing Yan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Jiangsu Province, China
| | - Qing Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - HuiTing Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Jiangsu Province, China
| | - YueTao Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - ShuQin Yu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Jiangsu Province, China
| | - GuangLin Xu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Jiangsu Province, China
| | - ZhiMin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Jiangsu Province, China
| | - ZhongJun Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- * E-mail: (ZL); (ZZ)
| | - ZhiHui Zhao
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Jiangsu Province, China
- * E-mail: (ZL); (ZZ)
| |
Collapse
|
28
|
Year in review 2010: Critical Care--Cardiology. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:241. [PMID: 22152086 PMCID: PMC3388636 DOI: 10.1186/cc10542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We review key research papers in cardiology and intensive care published during 2010 in Critical Care and quote related studies published in other journals if appropriate. Papers were grouped into the following categories: cardiovascular therapies, biomarkers, hemodynamic monitoring, cardiovascular diseases, and microcirculation.
Collapse
|
29
|
Neto HAP, Kubes P. Platelets, endothelium and shear join forces to mislead neutrophils in sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:103. [PMID: 21349145 PMCID: PMC3222017 DOI: 10.1186/cc9371] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neutrophils are circulating leukocytes with great cytotoxic potential, responsible for the first combat against invading pathogens. Their accumulation in tissues must be highly controlled so that the number of neutrophils delivered to the affected site is sufficient to control infection with minimum injury to the surrounding healthy tissue. In sepsis, neutrophil migration is dysregulated - resulting in insufficient delivery of neutrophils to the infectious site and massive neutrophil accumulation in uninfected organs. This dysregulation has the potential to cause inappropriate tissue injury that may explain the multiple organ dysfunction observed in severe sepsis. A better understanding of the mechanisms that contribute to this process is fundamental to design therapeutic strategies to circumvent tissue injury and organ dysfunction in sepsis.
Collapse
Affiliation(s)
- Heitor A Paula Neto
- Immunology Research Group, Department of Physiology and Pharmacology, Calvin, Phoebe & Joan Snyder Institute for Infection, Immunity & Inflammation, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | | |
Collapse
|