1
|
De Bruyn L, Vander Perre S, Verbruggen S, Joosten K, Van den Berghe G, Langouche L. Alterations in the lipid profile of critically ill children in relation to outcome. Crit Care 2025; 29:99. [PMID: 40038820 DOI: 10.1186/s13054-025-05327-5] [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: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Critically ill adults typically develop hypocholesterolemia, associated with poor outcome. Whether similar alterations occur in critically ill children is less clear. METHODS In secondary analyses of the PEPaNIC RCT (n = 1440), we first documented the time course of plasma cholesterol and triglyceride concentrations, and the effect of randomization to early-parenteral-nutrition (early-PN) or late-PN hereon, for 96 matched critically ill children staying ≥ 5 days in PICU. Second, for 1165 children with available admission plasma samples, lipid profiles were determined and their independent associations with outcome (time to live PICU discharge, new infection and 90-day mortality) were assessed with Multivariable Cox proportional hazard and logistic regression, adjusting for baseline risk factors. RESULTS Plasma HDL-cholesterol, LDL-cholesterol, total-cholesterol and triglycerides were low throughout the 5 PICU days, with only HDL-cholesterol further decreasing over time (P < 0.0001) and without effect of randomization to early-PN or late-PN, and with admission values lower in infants than older children and in patients with infection (P < 0.05). Lower admission HDL- and total-cholesterol concentrations were independently associated with a lower likelihood of an earlier live PICU discharge (P < 0.001) and with a higher risk of 90-day mortality (P ≤ 0.01), whereas higher plasma triglycerides were independently associated with higher risk of 90-day mortality (P = 0.004). Low admission plasma HDL-cholesterol was independently associated with a higher risk of acquiring a new infection (P = 0.05). CONCLUSION Critically ill children presented with low circulating levels of lipids. Low plasma cholesterol concentrations were associated with poor outcomes, most robustly for HDL-cholesterol. Whether these associations are causal or casual requires further investigation.
Collapse
Affiliation(s)
- Lauren De Bruyn
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Sarah Vander Perre
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Sascha Verbruggen
- Intensive Care Unit, Department of Neonatal and Paediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus Medical Centre-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Koen Joosten
- Intensive Care Unit, Department of Neonatal and Paediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus Medical Centre-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium.
| |
Collapse
|
2
|
Kumar P, Schroder EA, Rajaram MVS, Harris EN, Ganesan LP. The Battle of LPS Clearance in Host Defense vs. Inflammatory Signaling. Cells 2024; 13:1590. [PMID: 39329771 PMCID: PMC11430141 DOI: 10.3390/cells13181590] [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: 08/29/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Lipopolysaccharide (LPS) in blood circulation causes endotoxemia and is linked to various disease conditions. Current treatments focus on preventing LPS from interacting with its receptor Toll-like receptor 4 (TLR4) and reducing inflammation. However, our body has a natural defense mechanism: reticuloendothelial cells in the liver rapidly degrade and inactivate much of the circulating LPS within minutes. But this LPS clearance mechanism is not perfect. Excessive LPS that escape this clearance mechanism cause systemic inflammatory damage through TLR4. Despite its importance, the role of reticuloendothelial cells in LPS elimination is not well-studied, especially regarding the specific cells, receptors, and mechanisms involved. This gap hampers the development of effective therapies for endotoxemia and related diseases. This review consolidates the current understanding of LPS clearance, narrates known and explores potential mechanisms, and discusses the relationship between LPS clearance and LPS signaling. It also aims to highlight key insights that can guide the development of strategies to reduce circulating LPS by way of bolstering host defense mechanisms. Ultimately, we seek to provide a foundation for future research that could lead to innovative approaches for enhancing the body's natural ability to clear LPS and thereby lower the risk of endotoxin-related inflammatory diseases, including sepsis.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Evan A. Schroder
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA; (E.A.S.); (E.N.H.)
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Edward N. Harris
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA; (E.A.S.); (E.N.H.)
| | - Latha P. Ganesan
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
3
|
De Bruyn L, Téblick A, Van Oudenhove T, Vander Perre S, Derese I, Pauwels L, Derde S, De Vlieger G, Van den Berghe G, Langouche L. Glucocorticoid treatment increases cholesterol availability during critical illness: effect on adrenal and muscle function. Crit Care 2024; 28:295. [PMID: 39238038 PMCID: PMC11378467 DOI: 10.1186/s13054-024-05079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Hypocholesterolemia hallmarks critical illness though the underlying pathophysiology is incompletely understood. As low circulating cholesterol levels could partly be due to an increased conversion to cortisol/corticosterone, we hypothesized that glucocorticoid treatment, via reduced de novo adrenal cortisol/corticosterone synthesis, might improve cholesterol availability and as such affect adrenal gland and skeletal muscle function. METHODS In a matched set of prolonged critically ill patients (n = 324) included in the EPaNIC RCT, a secondary analysis was performed to assess the association between glucocorticoid treatment and plasma cholesterol from ICU admission to day five. Next, in a mouse model of cecal ligation and puncture-induced sepsis, septic mice were randomized to receive either hydrocortisone (1.2 mg/day) (n = 17) or placebo (n = 15) for 5 days, as compared with healthy mice (n = 18). Plasma corticosterone, cholesterol, and adrenocortical and myofiber cholesterol were quantified. Adrenal structure and steroidogenic capacity were evaluated. Muscle force and markers of atrophy, fibrosis and regeneration were quantified. In a consecutive mouse study with identical design (n = 24), whole body composition was assessed by EchoMRI to investigate impact on lean mass, fat mass, total and free water. RESULTS In human patients, glucocorticoid treatment was associated with higher plasma HDL- and LDL-cholesterol from respectively ICU day two and day three, up to day five (P < 0.05). Plasma corticosterone was no longer elevated in hydrocortisone-treated septic mice compared to placebo, whereas the sepsis-induced reduction in plasma HDL- and LDL-cholesterol and in adrenocortical cholesterol was attenuated (P < 0.05), but without improving the adrenocortical ACTH-induced CORT response and with increased adrenocortical inflammation and apoptosis (P < 0.05). Total body mass was further decreased in hydrocortisone-treated septic mice (P < 0.01) compared to placebo, with no additional effect on muscle mass, force or myofiber size. The sepsis-induced rise in markers of muscle atrophy and fibrosis was unaffected by hydrocortisone treatment, whereas markers of muscle regeneration were suppressed compared to placebo (P < 0.05). An increased loss of lean body mass and total and free water was observed in hydrocortisone-treated septic mice compared to placebo (P < 0.05). CONCLUSIONS Glucocorticoid treatment partially attenuated critical illness-induced hypocholesterolemia, but at a cost of impaired adrenal function, suppressed muscle regeneration and exacerbated loss of body mass.
Collapse
Affiliation(s)
- Lauren De Bruyn
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Arno Téblick
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Tim Van Oudenhove
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Sarah Vander Perre
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Inge Derese
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lies Pauwels
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Sarah Derde
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Greet De Vlieger
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium.
| |
Collapse
|
4
|
Deng H, Liang WY, Chen LQ, Yuen TH, Sahin B, Vasilescu DM, Trinder M, Walley K, Rensen PC, Boyd JH, Brunham LR. CETP inhibition enhances monocyte activation and bacterial clearance and reduces streptococcus pneumonia-associated mortality in mice. JCI Insight 2024; 9:e173205. [PMID: 38646937 PMCID: PMC11141867 DOI: 10.1172/jci.insight.173205] [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: 06/22/2023] [Accepted: 03/05/2024] [Indexed: 04/25/2024] Open
Abstract
Sepsis is a leading cause of mortality worldwide, and pneumonia is the most common cause of sepsis in humans. Low levels of high-density lipoprotein cholesterol (HDL-C) levels are associated with an increased risk of death from sepsis, and increasing levels of HDL-C by inhibition of cholesteryl ester transfer protein (CETP) decreases mortality from intraabdominal polymicrobial sepsis in APOE*3-Leiden.CETP mice. Here, we show that treatment with the CETP inhibitor (CETPi) anacetrapib reduced mortality from Streptococcus pneumoniae-induced sepsis in APOE*3-Leiden.CETP and APOA1.CETP mice. Mechanistically, CETP inhibition reduced the host proinflammatory response via attenuation of proinflammatory cytokine transcription and release. This effect was dependent on the presence of HDL, leading to attenuation of immune-mediated organ damage. In addition, CETP inhibition promoted monocyte activation in the blood prior to the onset of sepsis, resulting in accelerated macrophage recruitment to the lung and liver. In vitro experiments demonstrated that CETP inhibition significantly promoted the activation of proinflammatory signaling in peripheral blood mononuclear cells and THP1 cells in the absence of HDL; this may represent a mechanism responsible for improved bacterial clearance during sepsis. These findings provide evidence that CETP inhibition represents a potential approach to reduce mortality from pneumosepsis.
Collapse
Affiliation(s)
- Haoyu Deng
- Department of Medicine, Faculty of Medicine
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | - Wan Yi Liang
- Department of Microbiology and Immunology, Faculty of Science, and
| | - Le Qi Chen
- Department of Microbiology and Immunology, Faculty of Science, and
| | - Tin Ho Yuen
- Department of Microbiology and Immunology, Faculty of Science, and
| | - Basak Sahin
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | | | - Mark Trinder
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
- Department of Experimental Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Keith Walley
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | - Patrick C.N. Rensen
- Department of Medicine, Division of Endocrinology, and
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - John H. Boyd
- Department of Medicine, Faculty of Medicine
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| | - Liam R. Brunham
- Department of Medicine, Faculty of Medicine
- Centre for Heart and Lung Innovation, St. Paul’s Hospital
| |
Collapse
|
5
|
Zeng L, Tang H, Chen J, Deng Y, Zhao Y, Lei H, Wan Y, Pan Y, Deng Y. Causal association of lipoprotein-associated phospholipids on the risk of sepsis: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 14:1275132. [PMID: 38274232 PMCID: PMC10808779 DOI: 10.3389/fendo.2023.1275132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Background Many previous studies have revealed a close relationship between lipoprotein metabolism and sepsis, but their causal relationship has, until now, remained unclear. Therefore, we performed a two-sample Mendelian randomization analysis to estimate the causal relationship of lipoprotein-associated phospholipids with the risk of sepsis. Materials and methods A two-sample Mendelian randomization (MR) analysis was performed to investigate the causal relationship between lipoprotein-associated phospholipids and sepsis based on large-scale genome-wide association study (GWAS) summary statistics. MR analysis was performed using a variety of methods, including inverse variance weighted as the primary method, MR Egger, weighted median, simple mode, and weighted mode as complementary methods. Further sensitivity analyses were used to test the robustness of the data. Results After Bonferroni correction, the results of the MR analysis showed that phospholipids in medium high-density lipoprotein (HDL; ORIVW = 0.82, 95% CI 0.71-0.95, P = 0.0075), large HDL (ORIVW = 0.92, 95% CI 0.85-0.98, P = 0.0148), and very large HDL (ORMR Egger = 0.83, 95% CI 0.72-0.95, P = 0.0134) had suggestive causal relationship associations with sepsis. Sensitivity testing confirmed the accuracy of these findings. There was no clear association between other lipoprotein-associated phospholipids and sepsis risk. Conclusions Our MR analysis data suggestively showed a correlation between higher levels of HDL-associated phospholipids and reduced risk of sepsis. Further studies are required to determine the underlying mechanisms behind this relationship.
Collapse
Affiliation(s)
- Liying Zeng
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haoxuan Tang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiehai Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Anaesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Yijian Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunfeng Zhao
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hang Lei
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yufei Wan
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Pan
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yongqiang Deng
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Lauwers C, De Bruyn L, Langouche L. Impact of critical illness on cholesterol and fatty acids: insights into pathophysiology and therapeutic targets. Intensive Care Med Exp 2023; 11:84. [PMID: 38015312 PMCID: PMC10684846 DOI: 10.1186/s40635-023-00570-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
Critical illness is characterized by a hypercatabolic response encompassing endocrine and metabolic alterations. Not only the uptake, synthesis and metabolism of glucose and amino acids is majorly affected, but also the homeostasis of lipids and cholesterol is altered during acute and prolonged critical illness. Patients who suffer from critically ill conditions such as sepsis, major trauma, surgery or burn wounds display an immediate and sustained reduction in low plasma LDL-, HDL- and total cholesterol concentrations, together with a, less pronounced, increase in plasma free fatty acids. The severity of these alterations is associated with severity of illness, but the underlying pathophysiological mechanisms are multifactorial and only partly clarified. This narrative review aims to provide an overview of the current knowledge of how lipid and cholesterol uptake, synthesis and metabolism is affected during critical illness. Reduced nutritional uptake, increased scavenging of lipoproteins as well as an increased conversion to cortisol or other cholesterol-derived metabolites might all play a role in the decrease in plasma cholesterol. The acute stress response to critical illness creates a lipolytic cocktail, which might explain the increase in plasma free fatty acids, although reduced uptake and oxidation, but also increased lipogenesis, especially in prolonged critical illness, will also affect the circulating levels. Whether a disturbed lipid homeostasis warrants intervention or should primarily be interpreted as a signal of severity of illness requires further research.
Collapse
Affiliation(s)
- Caroline Lauwers
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lauren De Bruyn
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium.
| |
Collapse
|
7
|
Huang T, Fan Y, Xia Y, Xu X, Chen X, Ye H, Chen Y, Wang S. Association of low HDL-c levels with severe symptoms and poor clinical prognosis in patients with severe fever and thrombocytopenia syndrome. Front Microbiol 2023; 14:1239420. [PMID: 37720148 PMCID: PMC10501784 DOI: 10.3389/fmicb.2023.1239420] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Background Severe fever with thrombocytopenia syndrome (SFTS) is an acute infectious disease caused by a novel bunyavirus, characterized by high fever, thrombocytopenia, and multiple organ damage. Disturbances in lipid metabolism often occur during viral infections, but the changes and clinical significance of lipid profiles in SFTS patients remain unclear. This study aimed to investigate the alterations in lipid profiles and their clinical significance in SFTS patients. Methods A total of 157 SFTS patients and 157 healthy controls were enrolled in this study. Serum lipid levels were collected and analyzed among different groups and prognosis categories. Receiver operating characteristic (ROC) curve analysis was performed to assess the ability of lipid levels in distinguishing between severe and mild cases, as well as surviving and non-surviving patients. Pearson correlation analysis was used to examine the associations between lipid levels and clinical laboratory parameters. Results SFTS patients exhibited significantly lower levels of HDL-c, LDL-c, cholesterol, APoAI, and ApoB compared to healthy controls, while triglyceride levels were significantly higher. Serum HDL-c and ApoAI demonstrated good performance as indicators for distinguishing between survivors and non-survivors (AUC of 0.87 and 0.85, respectively). Multivariate regression analysis indicated that HDL-c independently acts as a protective factor in patients with SFTS. HDL-c levels showed decline in non-survivors but recovered in survivors. Moreover, HDL-c exhibited significant correlations with various clinical laboratory parameters (IL-6, CRP, AST, TT, APTT, PLT, ALB, and CD4). Conclusion This study identified abnormalities in serum lipid metabolism among SFTS patients. HDL-c and ApoAI levels hold potential as biomarkers for distinguishing survivors from non-survivors. Additionally, HDL-c and ApoAI may serve as therapeutic targets for the management of SFTS patients.
Collapse
Affiliation(s)
- Taihong Huang
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yinyin Fan
- Department of Pancreatic Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Yanyan Xia
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xuejing Xu
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xinyue Chen
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Hongling Ye
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuxin Chen
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Sen Wang
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Radford-Smith DE, Yates AG, Rizvi L, Anthony DC, Probert F. HDL and LDL have distinct, opposing effects on LPS-induced brain inflammation. Lipids Health Dis 2023; 22:54. [PMID: 37095493 PMCID: PMC10124044 DOI: 10.1186/s12944-023-01817-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
Endotoxemia and sepsis induce neuroinflammation and increase the risk of neurodegenerative disorders although the mechanism by which peripheral infection leads to brain inflammation is not well understood. While circulating serum lipoproteins are known immunometabolites with the potential to modulate the acute phase response and cross the blood brain barrier, their contribution to neuroinflammation during systemic infection is unknown. The objective of this study was to elucidate the mechanisms by which lipoprotein subclasses modulate lipopolysaccharide (LPS)-induced neuroinflammation. Adult C57BL/6 mice were divided into 6 treatment groups, including a sterile saline vehicle control group (n = 9), an LPS group (n = 11), a premixed LPS + HDL group (n = 6), a premixed LPS + LDL group (n = 5), a HDL only group (n = 6) and an LDL only group (n = 3). In all cases injections were administered intraperitoneally. LPS was administered at 0.5 mg/kg, and lipoproteins were administered at 20 mg/kg. Behavioural testing and tissue collection was performed 6 h post-injection. The magnitude of peripheral and central inflammation was determined by qPCR of pro-inflammatory genes in fresh liver and brain. Metabolite profiles of liver, plasma and brain were determined by 1H NMR. Endotoxin concentration in the brain was measured by the Limulus Amoebocyte Lysate (LAL) assay. Co-administration of LPS + HDL exacerbated both peripheral and central inflammation, whilst LPS + LDL attenuated this inflammation. Metabolomic analysis identified several metabolites significantly associated with LPS-induced inflammation, which were partially rescued by LDL, but not HDL. Endotoxin was detected at significantly greater concentrations in the brains of animals that received LPS + HDL compared to LPS + saline, but not those that received LPS + LDL. These results suggest that HDL may promote neuroinflammation through direct shuttling of endotoxin to the brain. In contrast, LDL was shown to have anti-neuroinflammatory properties in this study. Our results indicate that lipoproteins may be useful targets in neuroinflammation and neurodegeneration associated with endotoxemia and sepsis.
Collapse
Affiliation(s)
- Daniel E Radford-Smith
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK.
- Department of Chemistry, University of Oxford, Oxford, UK.
| | - Abi G Yates
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Laila Rizvi
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Daniel C Anthony
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Kargarpour Z, Panahipour L, Mildner M, Miron RJ, Gruber R. Lipids of Platelet-Rich Fibrin Reduce the Inflammatory Response in Mesenchymal Cells and Macrophages. Cells 2023; 12:cells12040634. [PMID: 36831301 PMCID: PMC9954017 DOI: 10.3390/cells12040634] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/18/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Platelet-rich fibrin (PRF) has a potent anti-inflammatory activity but the components mediating this effect remain unknown. Blood lipids have anti-inflammatory properties. The question arises whether this is also true for the lipid fraction of PRF. To answer this question, lipid fractions of solid and liquid PRF were tested for their potential to lower the inflammatory response of ST2 bone marrow stromal cells and primary bone marrow macrophages exposed to IL1β and TNFα, and LPS, respectively. Cytokine production and the underlying signalling pathway were analysed by RT-PCR, immunoassays, and Western blotting. We report here that lipids from solid and liquid PRF substantially lowered cytokine-induced expression of IL6, CCL2 and CCL5 in ST2 cells. Moreover, the inflammatory response induced by Pam3CSK4, the agonist of Toll-like receptor (TLR) TLR2, was partially reduced by the lipid extracts in ST2 cells. The PRF lipids further reduced the LPS-induced expression of IL1β, IL6 and CCL5 in macrophages at the transcriptional level. This was confirmed by showing the ability of PRF lipids to diminish IL6 at the protein level in ST2 cells and macrophages. Likewise, PRF lipid extracts reduced the phosphorylation of p38 and JNK and moderately decreased the phosphorylation of NFκB-p65 in ST2 cells. These findings suggest that the lipid fraction is at least partially responsible for the anti-inflammatory activity of PRF in vitro.
Collapse
Affiliation(s)
- Zahra Kargarpour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Department of Pulmonology, University Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Richard J. Miron
- Department of Periodontology, School of Dental Medicine, University of Bern, 3012 Bern, Switzerland
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Department of Periodontology, School of Dental Medicine, University of Bern, 3012 Bern, Switzerland
- Correspondence: ; Tel.: +43-1-40070-2660
| |
Collapse
|
10
|
Bonilha I, Luchiari B, Nadruz W, Sposito AC. Very low HDL levels: clinical assessment and management. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:3-18. [PMID: 36651718 PMCID: PMC9983789 DOI: 10.20945/2359-3997000000585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In individuals with very low high-density lipoprotein (HDL-C) cholesterol, such as Tangier disease, LCAT deficiency, and familial hypoalphalipoproteinemia, there is an increased risk of premature atherosclerosis. However, analyzes based on comparisons of populations with small variations in HDL-C mediated by polygenic alterations do not confirm these findings, suggesting that there is an indirect association or heterogeneity in the pathophysiological mechanisms related to the reduction of HDL-C. Trials that evaluated some of the HDL functions demonstrate a more robust degree of association between the HDL system and atherosclerotic risk, but as they were not designed to modify lipoprotein functionality, there is insufficient data to establish a causal relationship. We currently have randomized clinical trials of therapies that increase HDL-C concentration by various mechanisms, and this HDL-C elevation has not independently demonstrated a reduction in the risk of cardiovascular events. Therefore, this evidence shows that (a) measuring HDL-C as a way of estimating HDL-related atheroprotective system function is insufficient and (b) we still do not know how to increase cardiovascular protection with therapies aimed at modifying HDL metabolism. This leads us to a greater effort to understand the mechanisms of molecular action and cellular interaction of HDL, completely abandoning the traditional view focused on the plasma concentration of HDL-C. In this review, we will detail this new understanding and the new horizon for using the HDL system to mitigate residual atherosclerotic risk.
Collapse
Affiliation(s)
- Isabella Bonilha
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Beatriz Luchiari
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Wilson Nadruz
- Universidade de Campinas (Unicamp), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Andrei C Sposito
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil,
| |
Collapse
|
11
|
Abstract
Systemic inflammation has been suggested to have a pivotal role in atherothrombosis, but the factors that trigger systemic inflammation have not been fully elucidated. Lipopolysaccharide (LPS) is a component of the membrane of Gram-negative bacteria present in the gut that can translocate into the systemic circulation, causing non-septic, low-grade endotoxaemia. Gut dysbiosis is a major determinant of low-grade endotoxaemia via dysfunction of the intestinal barrier scaffold, which is a prerequisite for LPS translocation into the systemic circulation. Experimental studies have demonstrated that LPS is present in atherosclerotic arteries but not in normal arteries. In atherosclerotic plaques, LPS promotes a pro-inflammatory status that can lead to plaque instability and thrombus formation. Low-grade endotoxaemia affects several cell types, including leukocytes, platelets and endothelial cells, leading to inflammation and clot formation. Low-grade endotoxaemia has been described in patients at risk of or with overt cardiovascular disease, in whom low-grade endotoxaemia was associated with atherosclerotic burden and its clinical sequelae. In this Review, we describe the mechanisms favouring the development of low-grade endotoxaemia, focusing on gut dysbiosis and changes in gut permeability; the plausible biological mechanisms linking low-grade endotoxaemia and atherothrombosis; the clinical studies suggesting that low-grade endotoxaemia is a risk factor for cardiovascular events; and the potential therapeutic tools to improve gut permeability and eventually eliminate low-grade endotoxaemia.
Collapse
|
12
|
Lahoz C, Salinero-Fort MA, Cárdenas J, Rodríguez-Artalejo F, Díaz-Almiron M, Vich-Pérez P, San Andrés-Rebollo FJ, Vicente I, Mostaza JM. HDL-cholesterol concentration and risk of SARS-CoV-2 infection in people over 75 years of age: A cohort with half a million participants from the Community of Madrid. CLÍNICA E INVESTIGACIÓN EN ARTERIOSCLEROSIS (ENGLISH EDITION) 2022. [PMCID: PMC9132787 DOI: 10.1016/j.artere.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Objective The aim of this study was to analyze the relationship between HDL-cholesterol and the risk of SARS-CoV-2 infection in over 75-year-olds residing in the Community of Madrid. Methods Study of a population-based cohort, composed of all residents in Madrid (Spain) born before January 1, 1945 and alive on December 31, 2019. Demographic, clinical and analytical data were obtained from primary care electronic medical records from January 2015. Confirmed SARS-CoV-2 infection was defined as a positive RT-PCR or antigen test result. Infection data correspond to the period March 1, 2020 through December 31, 2020. Results Of the 593,342 cohort participants, 501,813 had at least one HDL-cholesterol determination in the past 5 years. Their mean age was 83.4 ± 5.6 years and 62.4% were women. A total of 36,996 (7.4%) had a confirmed SARS-CoV2 infection during 2020. The risk of infection [odds ratio (95% confidence interval)] for SARS-CoV2 according to increasing quintiles of HDL-cholesterol was 1, 0,960 (0,915–1,007), 0,891 (0,848–0,935), 0,865 (0,824–0,909) y 0,833 (0.792–0,876), after adjusting for age, sex, cardiovascular risk factors and comorbidities. Conclusions There is an inverse and dose-dependent relationship between HDL-cholesterol concentration and the risk of SARS-CoV2 infection in subjects aged over 75 years of age in the Community of Madrid.
Collapse
|
13
|
Lahoz C, Salinero-Fort MA, Cárdenas J, Rodríguez-Artalejo F, Díaz-Almiron M, Vich-Pérez P, San Andrés-Rebollo FJ, Vicente I, Mostaza JM. Concentración de colesterol-HDL y riesgo de infección por SARS-CoV-2 en personas mayores de 75 años: una cohorte con medio millón de participantes de la Comunidad de Madrid. CLÍNICA E INVESTIGACIÓN EN ARTERIOSCLEROSIS 2022; 34:113-119. [PMID: 35125250 PMCID: PMC8654578 DOI: 10.1016/j.arteri.2021.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/13/2021] [Accepted: 11/29/2021] [Indexed: 01/08/2023]
Abstract
Objetivo El objetivo de este estudio fue analizar la relación entre el colesterol-HDL y el riesgo de infección por SARS-CoV-2 en mayores de 75 años residentes en la Comunidad de Madrid. Métodos Estudio de una cohorte de base poblacional, compuesto por todos los residentes en Madrid (España) nacidos antes del 1 de enero de 1945 y vivos el 31 de diciembre de 2019. Los datos demográficos, clínicos y analíticos se obtuvieron de las historias clínicas electrónicas de atención primaria desde enero de 2015. La infección confirmada por SARS-CoV-2 se definió como un resultado positivo en la RT-PCR o en la prueba de antígeno. Los datos sobre infección por SARS-CoV-2 corresponden al periodo del 1 de marzo de 2020 hasta el 31 de diciembre de 2020. Resultados De los 593.342 participantes de la cohorte, 501.813 tenían al menos una determinación de colesterol-HDL en los últimos 5 años. Su edad media era 83,4 ± 5,6 años y el 62,4% eran mujeres. Un total de 36.996 (7,4%) tuvieron una infección confirmada por SARS-CoV2 durante el año 2020. El riesgo de infección (odds ratio [intervalo de confianza 95%]) por SARS-CoV2 según los quintiles crecientes de colesterol-HDL fue de 1; 0,960 (0,915-1,007), 0,891 (0,848-0,935), 0,865 (0,824-0,909) y 0,833 (0.792-0,876), tras ajustar por edad, sexo, factores de riesgo cardiovascular y comorbilidades. Conclusiones Existe una relación inversa y dosis-dependiente entre la concentración de colesterol-HDL y el riesgo de infección por SARS-CoV2 en los mayores de 75 años de la Comunidad de Madrid.
Collapse
|
14
|
Kingwell BA, Nicholls SJ, Velkoska E, Didichenko SA, Duffy D, Korjian S, Gibson CM. Antiatherosclerotic Effects of CSL112 Mediated by Enhanced Cholesterol Efflux Capacity. J Am Heart Assoc 2022; 11:e024754. [PMID: 35411789 PMCID: PMC9238469 DOI: 10.1161/jaha.121.024754] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Approximately 12% of patients with acute myocardial infarction (AMI) experience a recurrent major adverse cardiovascular event within 1 year of their primary event, with most occurring within the first 90 days. Thus, there is a need for new therapeutic approaches that address this 90-day post-AMI high-risk period. The formation and eventual rupture of atherosclerotic plaque that leads to AMI is elicited by the accumulation of cholesterol within the arterial intima. Cholesterol efflux, a mechanism by which cholesterol is removed from plaque, is predominantly mediated by apolipoprotein A-I, which is rapidly lipidated to form high-density lipoprotein in the circulation and has atheroprotective properties. In this review, we outline how cholesterol efflux dysfunction leads to atherosclerosis and vulnerable plaque formation, including inflammatory cell recruitment, foam cell formation, the development of a lipid/necrotic core, and degradation of the fibrous cap. CSL112, a human plasma-derived apolipoprotein A-I, is in phase 3 of clinical development and aims to reduce the risk of recurrent cardiovascular events in patients with AMI in the first 90 days after the index event by increasing cholesterol efflux. We summarize evidence from preclinical and clinical studies suggesting that restoration of cholesterol efflux by CSL112 can stabilize plaque by several anti-inflammatory/immune-regulatory processes. These effects occur rapidly and could stabilize vulnerable plaques in patients who have recently experienced an AMI, thereby reducing the risk of recurrent major adverse cardiovascular events in the high-risk early post-AMI period.
Collapse
Affiliation(s)
| | | | | | | | | | - Serge Korjian
- PERFUSE Study Group, Cardiovascular Division Departments of Medicine Beth Israel Deaconess Medical CenterHarvard Medical School Boston MA
| | - C Michael Gibson
- PERFUSE Study Group, Cardiovascular Division Departments of Medicine Beth Israel Deaconess Medical CenterHarvard Medical School Boston MA
| |
Collapse
|
15
|
Hong S, Niu M, Meng D, Li A, Dong Q, Zhang J, Tian X, Lu S, Wang Y. High-density lipoprotein reduces microglia activation and protects against experimental autoimmune encephalomyelitis in mice. Int Immunopharmacol 2022; 105:108566. [DOI: 10.1016/j.intimp.2022.108566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/05/2022]
|
16
|
Charó N, Jerez H, Tatti S, Romero EL, Schattner M. The Anti-Inflammatory Effect of Nanoarchaeosomes on Human Endothelial Cells. Pharmaceutics 2022; 14:736. [PMID: 35456570 PMCID: PMC9027062 DOI: 10.3390/pharmaceutics14040736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 01/14/2023] Open
Abstract
Archaebacterias are considered a unique source of novel biomaterials of interest for nanomedicine. In this perspective, the effects of nanoarchaeosomes (ARC), which are nanovesicles prepared from polar lipids extracted from the extreme halophilic Halorubrum tebenquinchense, on human umbilical vein endothelial cells (HUVEC) were investigated in physiological and under inflammatory static conditions. Upon incubation, ARC (170 nm mean size, -41 mV ζ) did not affect viability, cell proliferation, and expression of intercellular adhesion molecule-1 (ICAM-1) and E-selectin under basal conditions, but reduced expression of both molecules and secretion of IL-6 induced by lypopolysaccharide (LPS), Pam3CSK4 or Escherichia coli. Such effects were not observed with TNF-α or IL-1β stimulation. Interestingly, ARC significantly decreased basal levels of von Willebrand factor (vWF) and levels induced by all stimuli. None of these parameters was altered by liposomes of hydrogenated phosphatidylcholine and cholesterol of comparable size and concentration. Only ARC were endocytosed by HUVEC and reduced mRNA expression of ICAM-1 and vWF via NF-ĸB and ERK1/2 in LPS-stimulated cells. This is the first report of the anti-inflammatory effect of ARC on endothelial cells and our data suggest that its future use in vascular disease may hopefully be of particular interest.
Collapse
Affiliation(s)
- Nancy Charó
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina;
| | - Horacio Jerez
- Center for Research and Development in Nanomedicines (CIDEN), National University of Quilmes, Roque Saenz Peña, Bernal 1876, Argentina;
| | - Silvio Tatti
- Department of Obstetrics and Gynecology, Clinical Hospital, Av. Córdoba 2351, Buenos Aires 1120, Argentina;
| | - Eder Lilia Romero
- Center for Research and Development in Nanomedicines (CIDEN), National University of Quilmes, Roque Saenz Peña, Bernal 1876, Argentina;
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina;
| |
Collapse
|
17
|
Mostaza JM, Salinero-Fort MA, Cardenas-Valladolid J, Rodriguez-Artalejo F, Díaz-Almiron M, Vich-Pérez P, San Andres-Rebollo FJ, Vicente I, Lahoz C. Pre-infection HDL-cholesterol levels and mortality among elderly patients infected with SARS-CoV-2. Atherosclerosis 2021; 341:13-19. [PMID: 34959204 PMCID: PMC8692242 DOI: 10.1016/j.atherosclerosis.2021.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 01/28/2023]
Abstract
Background and aims Low HDL-cholesterol (HDLc) concentration is associated with a greater risk of infection-related mortality. We wanted to evaluate the relationship between pre-infection HDLc levels and mortality among older patients infected with SARS-Cov-2. Methods This is a population-based, cohort study, comprising all individuals residing in Madrid (Spain) born before 1 January 1945, and alive on 31 December 2019. Demographic, clinical, and analytical data were obtained from the primary care electronic clinical records. Confirmed SARS-CoV-2 infection was defined as a positive result in the RT-qPCR or in the antigen test. A death from COVID-19 was defined as that registered in the hospital chart, or as any death occurring in the 15 days following a confirmed SARS-CoV-2 infection. Data on infection, hospitalization, or death due to SAR-CoV-2 were collected from 1 March 2020 through 31 December 2020. Results Of the 593,342 individuals comprising the cohort, 36,966 had a SARS-CoV-2 infection during 2020, and at least one HDLc measurement in the previous five years. Among them, 9689 (26.2%) died from COVID-19. After adjustment for age and sex, the relative risk (95% confidence interval) of COVID-19 death across increasing quintiles of HDLc was 1.000, 0.896 (0.855–0.940), 0.816 (0.776–0.860), 0.758 (0.719–0.799), and 0.747 (0.708–0.787). The association was maintained after further adjustment for comorbidities, statin treatment and markers of malnutrition. While in females this association was linear, in males it showed a U-shaped curve. Conclusions In older subjects, a higher HDLc measured before SARS-CoV-2 infection was associated with a lower risk of death.
Collapse
Affiliation(s)
- Jose M Mostaza
- Lipid and Vascular Risk Unit, Department of Internal Medicine, Hospital Carlos III, Madrid, Spain.
| | - Miguel A Salinero-Fort
- Biosanitary Research and Innovation Foundation of Primary Care (FIIBAP), Hospital La Paz Institute for Health Research (IdIPAZ), Health Services Research on Chronic Patients Network (REDISSEC), Subdirectorate General for Health Research, Ministry of Health, Madrid, Spain
| | - Juan Cardenas-Valladolid
- Dirección Técnica de Sistemas de Información Sanitaria, Gerencia Adjunta de Procesos Asistenciales, Gerencia Asistencial de Atención Primaria, Madrid, Spain
| | - Fernando Rodriguez-Artalejo
- Department of Preventive Medicine and Public Health, Universidad Autónoma de Madrid-IdIPAZ, CIBER of Epidemiology and Public Health (CIBERESP) and IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Mariana Díaz-Almiron
- Research Unit, Hospital La Paz Institute for Health Research, IdiPAZ, Madrid, Spain
| | - Pilar Vich-Pérez
- Biosanitary Research and Innovation Foundation of Primary Care (FIIBAP), Los Alpes Health Center, Madrid, Spain
| | - F Javier San Andres-Rebollo
- Biosanitary Research and Innovation Foundation of Primary Care (FIIBAP), Los Alpes Health Center, Madrid, Spain
| | | | - Carlos Lahoz
- Lipid and Vascular Risk Unit, Department of Internal Medicine, Hospital Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Vestad B, Nyman TA, Hove-Skovsgaard M, Stensland M, Hoel H, Trøseid AMS, Aspelin T, Aass HCD, Puhka M, Hov JR, Nielsen SD, Øvstebø R, Trøseid M. Plasma extracellular vesicles in people living with HIV and type 2 diabetes are related to microbial translocation and cardiovascular risk. Sci Rep 2021; 11:21936. [PMID: 34754007 PMCID: PMC8578564 DOI: 10.1038/s41598-021-01334-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/22/2021] [Indexed: 11/09/2022] Open
Abstract
HIV and type 2 diabetes (T2D) are both associated with gut microbiota alterations, low-grade endotoxemia and increased cardiovascular risk. We investigated the potential role of plasma extracellular vesicles (EVs) in relation to these processes. Plasma EVs were isolated by size exclusion chromatography in fasting individuals with HIV and T2D (n = 16), T2D only (n = 14), HIV only (n = 20) or healthy controls (n = 19), and characterized by transmission electron microscopy, western blot, nanoparticle tracking analysis and quantitative proteomics. The findings were compared to gut microbiota alterations, lipopolysaccharide levels and cardiovascular risk profile. Individuals with concomitant HIV and T2D had higher plasma EV concentration, which correlated closely with plasma lipopolysaccharides, triglycerides and Framingham score, but not with gut microbiota alterations. Proteomic analyses identified 558 human proteins, largely related to cardiometabolic disease genes and upstream regulation of inflammatory pathways, including IL-6 and IL-1β, as well as 30 bacterial proteins, mostly from lipopolysaccharide-producing Proteobacteria. Our study supports that EVs are related to microbial translocation processes in individuals with HIV and T2D. Their proteomic content suggests a contributing role in low-grade inflammation and cardiovascular risk development. The present approach for exploring gut-host crosstalk can potentially identify novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Postboks 4590, 0424, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Norwegian Society for Extracellular Vesicles, NOR-EV, Oslo, Norway.
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Malene Hove-Skovsgaard
- Department of Infectious Diseases, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark
| | - Maria Stensland
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Hedda Hoel
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Postboks 4590, 0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Medical Department, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Anne-Marie Siebke Trøseid
- Norwegian Society for Extracellular Vesicles, NOR-EV, Oslo, Norway.,The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Trude Aspelin
- Norwegian Society for Extracellular Vesicles, NOR-EV, Oslo, Norway.,The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Hans Christian D Aass
- Norwegian Society for Extracellular Vesicles, NOR-EV, Oslo, Norway.,The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Maija Puhka
- Institute for Molecular Medicine Finland FIMM, EV and HiPrep Cores, University of Helsinki, Helsinki, Finland
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Postboks 4590, 0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Surgery, Inflammatory Medicine and Transplantation, Norwegian PSC Research Center and Section of Gastroenterology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Susanne Dam Nielsen
- Department of Infectious Diseases, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark
| | - Reidun Øvstebø
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Norwegian Society for Extracellular Vesicles, NOR-EV, Oslo, Norway.,The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Postboks 4590, 0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
19
|
Goossens C, Weckx R, Derde S, Vander Perre S, Derese I, Van Veldhoven PP, Ghesquière B, Van den Berghe G, Langouche L. Altered cholesterol homeostasis in critical illness-induced muscle weakness: effect of exogenous 3-hydroxybutyrate. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:252. [PMID: 34274000 PMCID: PMC8285799 DOI: 10.1186/s13054-021-03688-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022]
Abstract
Background Muscle weakness is a complication of critical illness which hampers recovery. In critically ill mice, supplementation with the ketone body 3-hydroxybutyrate has been shown to improve muscle force and to normalize illness-induced hypocholesterolemia. We hypothesized that altered cholesterol homeostasis is involved in development of critical illness-induced muscle weakness and that this pathway can be affected by 3-hydroxybutyrate. Methods In both human critically ill patients and septic mice, the association between circulating cholesterol concentrations and muscle weakness was assessed. In septic mice, the impact of 3-hydroxybutyrate supplementation on cholesterol homeostasis was evaluated with use of tracer technology and through analysis of markers of cholesterol metabolism and downstream pathways. Results Serum cholesterol concentrations were lower in weak than in non-weak critically ill patients, and in multivariable analysis adjusting for baseline risk factors, serum cholesterol was inversely correlated with weakness. In septic mice, plasma cholesterol correlated positively with muscle force. In septic mice, exogenous 3-hydroxybutyrate increased plasma cholesterol and altered cholesterol homeostasis, by normalization of plasma mevalonate and elevation of muscular, but not hepatic, expression of cholesterol synthesis genes. In septic mice, tracer technology revealed that 3-hydroxybutyrate was preferentially taken up by muscle and metabolized into cholesterol precursor mevalonate, rather than TCA metabolites. The 3-hydroxybutyrate protection against weakness was not related to ubiquinone or downstream myofiber mitochondrial function, whereas cholesterol content in myofibers was increased. Conclusions These findings point to a role for low cholesterol in critical illness-induced muscle weakness and to a protective mechanism-of-action for 3-hydroxybutyrate supplementation. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-021-03688-1.
Collapse
Affiliation(s)
- Chloë Goossens
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium
| | - Ruben Weckx
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium
| | - Sarah Derde
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium
| | - Sarah Vander Perre
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium
| | - Inge Derese
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium
| | - Paul P Van Veldhoven
- Laboratory for Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Center, Center for Cancer Biology, VIB, KU Leuven, 3000, Leuven, Belgium
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 bus 503, 3000, Leuven, Belgium.
| |
Collapse
|
20
|
Wang G, Deng J, Li J, Wu C, Dong H, Wu S, Zhong Y. The Role of High-Density Lipoprotein in COVID-19. Front Pharmacol 2021; 12:720283. [PMID: 34335279 PMCID: PMC8322438 DOI: 10.3389/fphar.2021.720283] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/06/2021] [Indexed: 01/08/2023] Open
Abstract
The current Coronavirus disease 2019 (COVID-19) pandemic has become a global challenge. Managing a large number of acutely ill patients in a short time, whilst reducing the fatality rate and dealing with complications, brings unique difficulties. The most striking pathophysiological features of patients with severe COVID-19 are dysregulated immune responses and abnormal coagulation function, which can result in multiple-organ failure and death. Normally metabolized high-density lipoprotein (HDL) performs several functions, including reverse cholesterol transport, direct binding to lipopolysaccharide (LPS) to neutralize LPS activity, regulation of inflammatory response, anti-thrombotic effects, antioxidant, and anti-apoptotic properties. Clinical data shows that significantly decreased HDL levels in patients with COVID-19 are correlated with both disease severity and mortality. However, the role of HDL in COVID-19 and its specific mechanism remain unclear. In this analysis, we review current evidence mainly in the following areas: firstly, the pathophysiological characteristics of COVID-19, secondly, the pleiotropic properties of HDL, thirdly, the changes and clinical significance of HDL in COVID-19, and fourthly the prospect of HDL-targeting therapy in COVID-19 to clarify the role of HDL in the pathogenesis of COVID-19 and discuss the potential of HDL therapy in COVID-19.
Collapse
Affiliation(s)
- Guyi Wang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinxiu Li
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenfang Wu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiyun Dong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shangjie Wu
- Department of Respiratory, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanjun Zhong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Jia C, Anderson JLC, Gruppen EG, Lei Y, Bakker SJL, Dullaart RPF, Tietge UJF. High-Density Lipoprotein Anti-Inflammatory Capacity and Incident Cardiovascular Events. Circulation 2021; 143:1935-1945. [PMID: 33840204 DOI: 10.1161/circulationaha.120.050808] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The role of high-density lipoprotein (HDL) function in cardiovascular disease represents an important emerging concept. The present study investigated whether HDL anti-inflammatory capacity is prospectively associated with first cardiovascular events in the general population. METHODS HDL anti-inflammatory capacity was determined as its ability to suppress TNFα (tumor necrosis factor α)-induced VCAM-1 (vascular cell adhesion molecule-1) mRNA expression in endothelial cells in vitro (results expressed as achieved percent reduction by individual HDL related to the maximum TNFα effect with no HDL present). In a nested case-control design of the PREVEND (Prevention of Renal and Vascular End Stage Disease) study, 369 cases experiencing a first cardiovascular event (combined end point of death from cardiovascular causes, ischemic heart disease, nonfatal myocardial infarction, and coronary revascularization) during a median of 10.5 years of follow-up were identified and individually matched to 369 controls with respect to age, sex, smoking status, and HDL cholesterol. Baseline samples were available in 340 cases and 340 matched controls. RESULTS HDL anti-inflammatory capacity was not correlated with HDL cholesterol or hsCRP (high-sensitivity C-reactive protein). HDL anti-inflammatory capacity was significantly lower in cases compared with controls (31.6% [15.7-44.2] versus 27.0% [7.4-36.1]; P<0.001) and was inversely associated with incident CVD in a fully adjusted model (odds ratio [OR] per 1 SD, 0.74 [CI, 0.61-0.90]; P=0.002). Furthermore, this association was approximately similar with all individual components of the cardiovascular disease end point. The HDL anti-inflammatory was not correlated with cholesterol efflux capacity (r=-0.02; P>0.05). When combining these 2 HDL function metrics in 1 model, both were significantly and independently associated with incident cardiovascular disease in a fully adjusted model (efflux: OR per 1 SD, 0.74; P=0.002; anti-inflammatory capacity: OR per 1 SD, 0.66; P<0.001). Adding HDL anti-inflammatory capacity improved risk prediction by the Framingham risk score, with a model likelihood-ratio statistic increase from 10.50 to 20.40 (P=0.002). CONCLUSIONS The HDL anti-inflammatory capacity, reflecting vascular protection against key steps in atherogenesis, was inversely associated with incident cardiovascular events in a general population cohort, independent of HDL cholesterol and HDL cholesterol efflux capacity. Adding HDL anti-inflammatory capacity to the Framingham risk score improves risk prediction.
Collapse
Affiliation(s)
- Congzhuo Jia
- Department of Pediatrics (C.J., J.L.C.A., U.J.F.T.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (C.J., Y.L., U.J.F.T.)
| | - Josephine L C Anderson
- Department of Pediatrics (C.J., J.L.C.A., U.J.F.T.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eke G Gruppen
- Department of Endocrinology, (E.G.G., R.P.F.D.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Nephrology (E.G.G., S.J.L.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (C.J., Y.L., U.J.F.T.)
| | - Stephan J L Bakker
- Department of Nephrology (E.G.G., S.J.L.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, (E.G.G., R.P.F.D.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics (C.J., J.L.C.A., U.J.F.T.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (C.J., Y.L., U.J.F.T.)
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden (U.J.F.T.)
| |
Collapse
|
22
|
Dusuel A, Deckert V, Pais de Barros JP, van Dongen K, Choubley H, Charron É, Le Guern N, Labbé J, Mandard S, Grober J, Lagrost L, Gautier T. Human cholesteryl ester transfer protein lacks lipopolysaccharide transfer activity, but worsens inflammation and sepsis outcomes in mice. J Lipid Res 2020; 62:100011. [PMID: 33500240 PMCID: PMC7859855 DOI: 10.1194/jlr.ra120000704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 11/25/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
Bacterial lipopolysaccharides (LPSs or endotoxins) can bind most proteins of the lipid transfer/LPS-binding protein (LT/LBP) family in host organisms. The LPS-bound LT/LBP proteins then trigger either an LPS-induced proinflammatory cascade or LPS binding to lipoproteins that are involved in endotoxin inactivation and detoxification. Cholesteryl ester transfer protein (CETP) is an LT/LBP member, but its impact on LPS metabolism and sepsis outcome is unclear. Here, we performed fluorescent LPS transfer assays to assess the ability of CETP to bind and transfer LPS. The effects of intravenous (iv) infusion of purified LPS or polymicrobial infection (cecal ligation and puncture [CLP]) were compared in transgenic mice expressing human CETP and wild-type mice naturally having no CETP activity. CETP displayed no LPS transfer activity in vitro, but it tended to reduce biliary excretion of LPS in vivo. The CETP expression in mice was associated with significantly lower basal plasma lipid levels and with higher mortality rates in both models of endotoxemia and sepsis. Furthermore, CETPTg plasma modified cytokine production of macrophages in vitro. In conclusion, despite having no direct LPS binding and transfer property, human CETP worsens sepsis outcomes in mice by altering the protective effects of plasma lipoproteins against endotoxemia, inflammation, and infection.
Collapse
Affiliation(s)
- Aloïs Dusuel
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Valérie Deckert
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Jean-Paul Pais de Barros
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Kevin van Dongen
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Hélène Choubley
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Émilie Charron
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Naig Le Guern
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Jérôme Labbé
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Stéphane Mandard
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Jacques Grober
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Laurent Lagrost
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France; University Hospital of Dijon, Dijon, France
| | - Thomas Gautier
- INSERM/University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France.
| |
Collapse
|
23
|
Han Q, Han Y, Wen H, Pang Y, Li Q. Molecular Evolution of Apolipoprotein Multigene Family and the Original Functional Properties of Serum Apolipoprotein (LAL2) in Lampetra japonica. Front Immunol 2020; 11:1751. [PMID: 32849624 PMCID: PMC7431520 DOI: 10.3389/fimmu.2020.01751] [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: 02/25/2020] [Accepted: 06/30/2020] [Indexed: 01/20/2023] Open
Abstract
Apolipoprotein (APO) genes represent a large family of genes encoding various binding proteins associated with plasma lipid transport. Due to the long divergence history, it remains to be confirmed whether these genes evolved from a common ancestor through gene duplication and original function, and how this evolution occurred. In this study, based on the phylogenetic tree, sequence alignment, motifs, and evolutionary analysis of gene synteny and collinearity, APOA, APOC, and APOE in higher vertebrates may have a common ancestor, lamprey serum apolipoprotein LAL1 or LAL2, which traces back to 360 million years ago. Moreover, the results of immunofluorescence, immunohistochemistry, and flow cytometry show that LAL2 is primarily distributed in the liver, kidney, and blood leukocytes of lampreys, and specifically localized in the cytoplasm of liver cells and leukocytes, as well as secreted into sera. Surface plasmon resonance technology demonstrates that LAL2 colocalizes to breast adenocarcinoma cells (MCF-7) or chronic myeloid leukemia cells (K562) associated with lamprey immune protein (LIP) and further enhances the killing effect of LIP on tumor cells. In addition, using quantitative real-time PCR (Q-PCR) and western blot methods, we found that the relative mRNA and protein expression of lal2 in lamprey leukocytes and sera increased significantly at different times after stimulating with Staphylococcus aureus, Vibrio anguillarum, and Polyinosinic-polycytidylic acid (Poly I:C). Moreover, LAL2 was found to recognize and bind to gram-positive bacteria (Staphylococcus aureus and Bacillus cereus) and gram-negative bacteria (Escherichia coli) and play an important role in the antibacterial process. All in all, our data reveals a long, complex evolutionary history for apolipoprotein genes under different selection pressures, confirms the immune effect of LAL2 in lamprey sera against pathogens, and lays the foundation for further research regarding biological functions of lamprey immune systems.
Collapse
Affiliation(s)
- Qing Han
- College of Life Sciences, Liaoning Normal University, Dalian, China.,Lamprey Research Center, Liaoning Normal University, Dalian, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yinglun Han
- College of Life Sciences, Liaoning Normal University, Dalian, China.,Lamprey Research Center, Liaoning Normal University, Dalian, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Hongyan Wen
- College of Life Sciences, Liaoning Normal University, Dalian, China.,Lamprey Research Center, Liaoning Normal University, Dalian, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yue Pang
- College of Life Sciences, Liaoning Normal University, Dalian, China.,Lamprey Research Center, Liaoning Normal University, Dalian, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Qingwei Li
- College of Life Sciences, Liaoning Normal University, Dalian, China.,Lamprey Research Center, Liaoning Normal University, Dalian, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
24
|
Blauw LL, Wang Y, Willems van Dijk K, Rensen PCN. A Novel Role for CETP as Immunological Gatekeeper: Raising HDL to Cure Sepsis? Trends Endocrinol Metab 2020; 31:334-343. [PMID: 32033866 DOI: 10.1016/j.tem.2020.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/11/2019] [Accepted: 01/09/2020] [Indexed: 12/26/2022]
Abstract
Raising HDL using cholesteryl ester transfer protein (CETP) inhibitors failed to show a clinically relevant risk reduction of cardiovascular disease in clinical trials, inviting reconsideration of the role of CETP and HDL in human physiology. Based on solid evidence from studies with isolated macrophages, rodents, and humans, we propose that a major function of CETP may be to modulate HDL in order to help resolve bacterial infections. When gram-negative bacteria invade the blood, as occurs in sepsis, Kupffer cells lose their expression of CETP to increase HDL levels. This rise in HDL prevents systemic endotoxemia by binding lipopolysaccharide and induces a systemic proinflammatory response in macrophages to mediate bacterial clearance. This raises the interesting possibility to repurpose CETP inhibitors for the treatment of sepsis.
Collapse
Affiliation(s)
- Lisanne L Blauw
- Department of Internal Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Yanan Wang
- Department of Internal Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
25
|
Basauri A, González-Fernández C, Fallanza M, Bringas E, Fernandez-Lopez R, Giner L, Moncalián G, de la Cruz F, Ortiz I. Biochemical interactions between LPS and LPS-binding molecules. Crit Rev Biotechnol 2020; 40:292-305. [DOI: 10.1080/07388551.2019.1709797] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Arantza Basauri
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | | | - Marcos Fallanza
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | - Eugenio Bringas
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | - Raúl Fernandez-Lopez
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Laura Giner
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Gabriel Moncalián
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Fernando de la Cruz
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Inmaculada Ortiz
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
26
|
Alharbi A, Thompson JP, Brindle NP, Stover CM. Ex vivo modelling of the formation of inflammatory platelet-leucocyte aggregates and their adhesion on endothelial cells, an early event in sepsis. Clin Exp Med 2019; 19:321-337. [PMID: 30191349 PMCID: PMC6647484 DOI: 10.1007/s10238-018-0526-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/27/2018] [Indexed: 12/18/2022]
Abstract
Septicaemia is an acute inflammatory reaction in the bloodstream to the presence of pathogen-associated molecular patterns. Whole blood stimulation assays capture endotoxin-induced formation of aggregates between platelets and leucocytes using flow cytometry. We wanted to assess extent of spontaneous aggregate formation in whole blood stimulation assays and compare the effects of endotoxin and heat-killed, clinically relevant, bacterial pathogens on aggregate formation and then on adhesion of aggregates to TNFα-stimulated endothelial cells. We found that endotoxin (from Escherichia coli or Salmonella enteritidis) was not a suitable stimulus to provoke platelet-leucocyte aggregates in vitro, as it did not further increase the extent of aggregates formed spontaneously in stasis of hirudin-anticoagulated blood. Specifically, whole blood samples stimulated with or without LPS produced aggregates with a mean surface area of 140.97 and 117.68 μm2, respectively. By contrast, incubation of whole blood with heat-killed Klebsiella pneumoniae or Staphylococcus aureus produced significantly enhanced and complex cellular aggregates (with a mean surface area of 470.61 and 518.39 μm2, respectively) which adhered more frequently to TNFα (and free fatty acid)-stimulated endothelial cells. These were reliably captured by scanning electron microscopy. Adhesion of cellular aggregates could be blocked by incubation of endothelial cells with a commercial P-selectin antibody and an angiopoietin-2 ligand trap. In conclusion, we have developed an in vitro method that models the acute inflammatory reaction in whole blood in the presence of sepsis-relevant bacterial pathogen surfaces.
Collapse
Affiliation(s)
- Azzah Alharbi
- Department of Infection, Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jonathan P Thompson
- Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - Nicholas P Brindle
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
- Department of Molecular & Cell Biology, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
27
|
Trinder M, Boyd JH, Brunham LR. Molecular regulation of plasma lipid levels during systemic inflammation and sepsis. Curr Opin Lipidol 2019; 30:108-116. [PMID: 30649022 DOI: 10.1097/mol.0000000000000577] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Sepsis is a common syndrome of multiorgan system dysfunction caused by a dysregulated inflammatory response to an infection and is associated with high rates of mortality. Plasma lipid and lipoprotein levels and composition change profoundly during sepsis and have emerged as both biomarkers and potential therapeutic targets for this condition. The purpose of this article is to review recent progress in the understanding of the molecular regulation of lipid metabolism during sepsis. RECENT FINDINGS Patients who experience greater declines in high-density lipoprotein during sepsis are at much greater risk of succumbing to organ failure and death. Although the causality of these findings remains unclear, all lipoprotein classes can sequester and prevent the excessive inflammation caused by pathogen-associated lipids during severe infections such as sepsis. This primordial innate immune function has been best characterized for high-density lipoproteins. Most importantly, results from human genetics and preclinical animal studies have suggested that several lipid treatment strategies, initially designed for atherosclerosis, may hold promise as therapies for sepsis. SUMMARY Lipid and lipoprotein metabolism undergoes significant changes during sepsis. An improved understanding of the molecular regulation of these changes may lead to new opportunities for the treatment of sepsis.
Collapse
Affiliation(s)
- Mark Trinder
- Centre for Heart Lung Innovation
- Department of Experimental Medicine Program
| | - John H Boyd
- Centre for Heart Lung Innovation
- Department of Experimental Medicine Program
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liam R Brunham
- Centre for Heart Lung Innovation
- Department of Experimental Medicine Program
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
28
|
Kajani S, Curley S, McGillicuddy FC. Unravelling HDL-Looking beyond the Cholesterol Surface to the Quality Within. Int J Mol Sci 2018; 19:ijms19071971. [PMID: 29986413 PMCID: PMC6073561 DOI: 10.3390/ijms19071971] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/03/2018] [Accepted: 07/04/2018] [Indexed: 12/11/2022] Open
Abstract
High-density lipoprotein (HDL) particles have experienced a turbulent decade of falling from grace with widespread demotion from the most-sought-after therapeutic target to reverse cardiovascular disease (CVD), to mere biomarker status. HDL is slowly emerging from these dark times due to the HDL flux hypothesis wherein measures of HDL cholesterol efflux capacity (CEC) are better predictors of reduced CVD risk than static HDL-cholesterol (HDL-C) levels. HDL particles are emulsions of metabolites, lipids, protein, and microRNA (miR) built on the backbone of Apolipoprotein A1 (ApoA1) that are growing in their complexity due to the higher sensitivity of the respective “omic” technologies. Our understanding of particle composition has increased dramatically within this era and has exposed how our understanding of these particles to date has been oversimplified. Elucidation of the HDL proteome coupled with the identification of specific miRs on HDL have highlighted the “hormonal” characteristics of HDL in that it carries and delivers messages systemically. HDL can dock to most peripheral cells via its receptors, including SR-B1, ABCA1, and ABCG1, which may be a critical step for facilitating HDL-to-cell communication. The composition of HDL particles is, in turn, altered in numerous disease states including diabetes, auto-immune disease, and CVD. The consequence of changes in composition, however, on subsequent biological activities of HDL is currently poorly understood and this is an important avenue for the field to explore in the future. Improving HDL particle quality as opposed to HDL quantity may, in turn, prove a more beneficial investment to reduce CVD risk.
Collapse
Affiliation(s)
- Sarina Kajani
- Cardiometabolic Research Group, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, 4 Dublin, Ireland.
| | - Sean Curley
- Cardiometabolic Research Group, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, 4 Dublin, Ireland.
| | - Fiona C McGillicuddy
- Cardiometabolic Research Group, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, 4 Dublin, Ireland.
| |
Collapse
|
29
|
Wei XB, Chen XJ, Li YL, Huang JL, Chen XL, Yu DQ, Tan N, Liu YH, Chen JY, He PC. Apolipoprotein A-I: A favorable prognostic marker in infective endocarditis. J Clin Lipidol 2017; 12:498-505. [PMID: 29339066 DOI: 10.1016/j.jacl.2017.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/10/2017] [Accepted: 12/12/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND Decreased apolipoprotein A-I (apoA-I) and high-density lipoprotein cholesterol (HDL-C) are common in inflammation and sepsis. No study with a large sample size has been performed to investigate the prognostic value of apoA-I or HDL-C in infective endocarditis (IE). OBJECTIVE The present study aimed to explore the prognostic value of apoA-I and HDL-C for adverse outcomes in IE patients. METHODS Patients with a definite diagnosis of IE between January 2009 and July 2015 were enrolled and divided into 3 groups according to their apoA-I tertiles at admission. Univariate and multivariate analyses were performed to evaluate the relationship of apoA-I and HDL-C with clinical outcomes. RESULTS Of the 593 included patients, 40 (6.7%) died in hospital. Patients with lower apoA-I experienced markedly higher rates of in-hospital mortality (10.7%, 7.0%, and 2.5% in tertiles 1-3, respectively; P = .006) and major adverse clinical events (32.5%, 24.1%, and 8.6% in tertiles 1-3, respectively; P < .001). ApoA-I (area under the curve, 0.671; P < .001) and HDL-C (area under the curve, 0.672; P < .001) had predictive values for in-hospital death. Multivariate logistic regression showed that apoA-I <0.90 g/L and HDL-C <0.78 mmol/L were independent risk predictors for in-hospital death. A multivariate Cox proportional hazard analysis revealed that apoA-I (increments of 1 g/L; hazard ratio, 0.36; 95% confidence interval, 0.15-0.87; P = .023) and HDL-C (increments of 1 mmol/L; hazard ratio, 0.38; 95% confidence interval, 0.18-0.83; P = .015) were independently associated with long-term mortality. CONCLUSIONS ApoA-I and HDL-C were inversely associated with adverse IE prognosis.
Collapse
Affiliation(s)
- Xue-Biao Wei
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Xiao-Jin Chen
- Department of Internal Medicine, Longnan Medicine Hospital, Ganzhou, China
| | - Yuan-Ling Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Jie-Leng Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Xiao-Lan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Dan-Qing Yu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Yuan-Hui Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China.
| | - Ji-Yan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China.
| | - Peng-Cheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China; Department of Cardiology, The Second People's Hospital of Nanhai District, Guangdong General Hospital's Nanhai Hospital, Foshan, China.
| |
Collapse
|
30
|
Emerging role of various signaling pathways in the pathogenesis and therapeutics of atherosclerosis. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.rvm.2017.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
Rouer M, Alsac JM, Louedec L, Shoukr FA, Rouzet F, Michel JB, Meilhac O, Delbosc S. High-density lipoprotein therapy inhibits Porphyromonas gingivalis-induced abdominal aortic aneurysm progression. Thromb Haemost 2017; 115:789-99. [DOI: 10.1160/th15-05-0398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 11/13/2015] [Indexed: 12/19/2022]
Abstract
SummaryClinical and experimental studies have highlighted the potential implication of periondontal bacteria contamination in the pathogenesis of abdominal aortic aneurysms (AAA). In addition to their role in reverse cholesterol transport, high-density lipoproteins (HDLs) display multiple functions, including anti-inflammatory and lipopolysaccharide scavenging properties. Low plasma levels of HDL-cholesterol have been reported in AAA patients. We tested the effect of a HDL therapy in Sprague-Dawley rat model of AAA, obtained by intraluminal elastase infusion followed by repeated injections of Porphyromonas gingivalis (Pg). HDLs, isolated by ultracentrifugation of plasma from healthy human volunteers, were co-injected intravenously (10 mg/kg) with Pg (1.107 Colony Forming Unit) one, eight and 15 days after elastase perfusion. Rats were sacrificed one week after the last injection. Our results show that Pg injections promote the formation of a persistent neutrophil-rich thrombus associated with increased aortic diameter in this AAA model. HDLs significantly reduced the increased AAA diameter induced by Pg. Histology showed the onset of a healing process in the Pg/HDL group. HDL injections also reduced neutrophil activation in Pg-injected rats associated with decreased cytokine levels in conditioned media and plasma. Scintigraphic analysis showed an intense uptake of 99mTc-HDL by the AAA suggesting that HDLs could exert their beneficial effect by acting directly on the thrombus components. HDL supplementation may therefore constitute a new therapeutic tool for AAA treatment.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|
32
|
Zannis VI, Su S, Fotakis P. Role of apolipoproteins, ABCA1 and LCAT in the biogenesis of normal and aberrant high density lipoproteins. J Biomed Res 2017; 31:471. [PMID: 29109329 PMCID: PMC6307667 DOI: 10.7555/jbr.31.20160082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/30/2016] [Indexed: 12/28/2022] Open
Abstract
In this review, we focus on the pathway of biogenesis of HDL, the essential role of apoA-I, ATP binding cassette transporter A1 (ABCA1), and lecithin: cholesterol acyltransferase (LCAT) in the formation of plasma HDL; the generation of aberrant forms of HDL containing mutant apoA-I forms and the role of apoA-IV and apoE in the formation of distinct HDL subpopulations. The biogenesis of HDL requires functional interactions of the ABCA1 with apoA-I (and to a lesser extent with apoE and apoA-IV) and subsequent interactions of the nascent HDL species thus formed with LCAT. Mutations in apoA-I, ABCA1 and LCAT either prevent or impair the formation of HDL and may also affect the functionality of the HDL species formed. Emphasis is placed on three categories of apoA-I mutations. The first category describes a unique bio-engineered apoA-I mutation that disrupts interactions between apoA-I and ABCA1 and generates aberrant preβ HDL subpopulations that cannot be converted efficiently to α subpopulations by LCAT. The second category describes natural and bio-engineered apoA-I mutations that generate preβ and small size α4 HDL subpopulations, and are associated with low plasma HDL levels. These phenotypes can be corrected by excess LCAT. The third category describes bio-engineered apoA-I mutations that induce hypertriglyceridemia that can be corrected by excess lipoprotein lipase and also have defective maturation of HDL. The HDL phenotypes described here may serve in the future for diagnosis, prognoses and potential treatment of abnormalities that affect the biogenesis and functionality of HDL.
Collapse
Affiliation(s)
- Vassilis I. Zannis
- . Molecular Genetics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
- . Department University of Crete, School of Medicine, Heraklion, Crete, Greece
| | - Shi Su
- . Molecular Genetics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Panagiotis Fotakis
- . Molecular Genetics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
- . Department University of Crete, School of Medicine, Heraklion, Crete, Greece
| |
Collapse
|
33
|
Dysfunctional HDL in diabetes mellitus and its role in the pathogenesis of cardiovascular disease. Mol Cell Biochem 2017; 440:167-187. [PMID: 28828539 DOI: 10.1007/s11010-017-3165-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/16/2017] [Indexed: 12/17/2022]
Abstract
Coronary artery disease, the leading cause of death in the developed and developing countries, is prevalent in diabetes mellitus with 68% cardiovascular disease (CVD)-related mortality. Epidemiological studies suggested inverse correlation between HDL and CVD occurrence. Therefore, low HDL concentration observed in diabetic patients compared to non-diabetic individuals was thought to be one of the primary causes of increased risks of CVD. Efforts to raise HDL level via CETP inhibitors, Torcetrapib and Dalcetrapib, turned out to be disappointing in outcome studies despite substantial increases in HDL-C, suggesting that factors beyond HDL concentration may be responsible for the increased risks of CVD. Therefore, recent studies have focused more on HDL function than on HDL levels. The metabolic environment in diabetes mellitus condition such as hyperglycemia-induced advanced glycation end products, oxidative stress, and inflammation promote HDL dysfunction leading to greater risks of CVD. This review discusses dysfunctional HDL as one of the mechanisms of increased CVD risks in diabetes mellitus through adversely affecting components that support HDL function in cholesterol efflux and LDL oxidation. The dampening of reverse cholesterol transport, a key process that removes cholesterol from lipid-laden macrophages in the arterial wall, leads to increased risks of CVD in diabetic patients. Therapeutic approaches to keep diabetes under control may benefit patients from developing CVD.
Collapse
|
34
|
Dietary Milk Sphingomyelin Reduces Systemic Inflammation in Diet-Induced Obese Mice and Inhibits LPS Activity in Macrophages. BEVERAGES 2017. [DOI: 10.3390/beverages3030037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Opacification Domain of Serum Opacity Factor Inhibits Beta-Hemolysis and Contributes to Virulence of Streptococcus pyogenes. mSphere 2017; 2:mSphere00147-17. [PMID: 28435893 PMCID: PMC5397570 DOI: 10.1128/mspheredirect.00147-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/01/2017] [Indexed: 11/25/2022] Open
Abstract
Streptococcus pyogenes is a major human pathogen causing more than 700 million infections annually. As a successful pathogen, S. pyogenes produces many virulence factors that facilitate colonization, proliferation, dissemination, and tissue damage. Serum opacity factor (SOF), an extracellular protein, is one of the virulence factors made by S. pyogenes. The underlying mechanism of how SOF contributes to virulence is not fully understood. SOF has two major features: (i) it opacifies host serum by interacting with high-density lipoprotein, and (ii) it inhibits beta-hemolysis on blood agar. In this study, we demonstrate that the domain of SOF essential for opacifying serum is also essential for SOF-mediated beta-hemolysis inhibition and SOF-mediated virulence. Our results shed new light on the molecular mechanisms of SOF-host interaction. Serum opacity factor (SOF) is a cell surface virulence factor made by the human pathogen Streptococcus pyogenes. We found that S. pyogenes strains with naturally occurring truncation mutations in the sof gene have markedly enhanced beta-hemolysis. Moreover, deletion of the sof gene in a SOF-positive parental strain resulted in significantly increased beta-hemolysis. Together, these observations suggest that SOF is an inhibitor of beta-hemolysis. SOF has two major functional domains, including an opacification domain and a fibronectin-binding domain. Using a SOF-positive serotype M89 S. pyogenes parental strain and a panel of isogenic mutant derivative strains, we evaluated the relative contribution of each SOF functional domain to beta-hemolysis inhibition and bacterial virulence. We found that the opacification domain, rather than the fibronectin-binding domain, is essential for SOF-mediated beta-hemolysis inhibition. The opacification domain, but not the fibronectin-binding domain of SOF, also contributed significantly to virulence in mouse models of bacteremia and necrotizing myositis. Inasmuch as the opacification domain of SOF is known to interact avidly with host high-density lipoprotein (HDL), we speculate that SOF-HDL interaction is an important process underlying SOF-mediated beta-hemolysis inhibition and SOF-mediated virulence. IMPORTANCEStreptococcus pyogenes is a major human pathogen causing more than 700 million infections annually. As a successful pathogen, S. pyogenes produces many virulence factors that facilitate colonization, proliferation, dissemination, and tissue damage. Serum opacity factor (SOF), an extracellular protein, is one of the virulence factors made by S. pyogenes. The underlying mechanism of how SOF contributes to virulence is not fully understood. SOF has two major features: (i) it opacifies host serum by interacting with high-density lipoprotein, and (ii) it inhibits beta-hemolysis on blood agar. In this study, we demonstrate that the domain of SOF essential for opacifying serum is also essential for SOF-mediated beta-hemolysis inhibition and SOF-mediated virulence. Our results shed new light on the molecular mechanisms of SOF-host interaction.
Collapse
|
36
|
van Bergenhenegouwen J, Kraneveld AD, Rutten L, Garssen J, Vos AP, Hartog A. Lipoproteins attenuate TLR2 and TLR4 activation by bacteria and bacterial ligands with differences in affinity and kinetics. BMC Immunol 2016; 17:42. [PMID: 27793087 PMCID: PMC5086051 DOI: 10.1186/s12865-016-0180-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/19/2016] [Indexed: 12/28/2022] Open
Abstract
Background The small intestine is a specialized compartment were close interactions take place between host, microbes, food antigens and dietary fatty acids. Dietary fats get absorbed by epithelial cells and processed into a range of lipoprotein particles after which they are basolaterally secreted and collected in the lymphatics. In contrast to the colon, the small intestine is covered only by a thin mucus coat that allows for intimate interactions between host-cells and microbes. Lipoproteins have long been recognized as protective factors in infectious diseases via the neutralization of bacterial toxins like lipopolysaccharides. Much less attention has been given to the potential role of lipoproteins as factors contributing to the maintenance of small intestinal immune homeostasis via modulating bacteria-induced immune responses. Results Lipoproteins VLDL, LDL and HDL were found to neutralize TLR responses towards specific TLR-ligands or a selection of gram-negative and gram-positive bacteria. Attenuation of TLR2 activity was acute and only slightly improved by longer pre-incubation times of ligands and lipoproteins with no differences between bacterial-lipopeptides or bacteria. In contrast, attenuation of TLR4 responses was only observed after extensive preincubation of lipoproteins and LPS. Preincubation of bacteria and lipoproteins led only to a modest attenuation of TLR4 activity. Moreover, compared to TLR2, TLR4 activity could only be attenuated by lipoproteins over a small ligand dose range. Conclusions These results demonstrate the ability of lipoproteins VLDL, LDL and HDL to inhibit TLR responses towards bacterial-ligands and bacteria. Presence of lipoproteins was found to modulate the MAMP-induced cytokine release by primary human monocytes measured as changes in the release of IL-6, TNFα, GM-CSF and IFNγ. Using TLR2 and TLR4-reporter cells, lipoproteins were found to inhibit TLR responses with differences in affinity and kinetics. These data establish a role for lipoproteins as immunoregulatory molecules, attenuating TLR-responses and thereby positively contributing to mucosal homeostasis. Electronic supplementary material The online version of this article (doi:10.1186/s12865-016-0180-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeroen van Bergenhenegouwen
- Nutricia Research, Utrecht, The Netherlands. .,Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| | - Aletta D Kraneveld
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Johan Garssen
- Nutricia Research, Utrecht, The Netherlands.,Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Anita Hartog
- Nutricia Research, Utrecht, The Netherlands.,Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
37
|
Berbee JF, Havekes LM, Rensen PC. Apolipoproteins modulate the inflammatory response to lipopolysaccharide. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110020501] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An increasing body of evidence demonstrates a close interplay between lipoprotein metabolism and sepsis. Sepsis results in an increase of plasma triglycerides within VLDL as a consequence of an enhanced hepatic VLDL production and/or inhibited peripheral and hepatic VLDL clearance. In contrast, sepsis decreases plasma cholesterol within LDL and mainly HDL. The decrease in HDL is accompanied by a loss of mainly apoAI-containing particles, an almost total loss of apoCI, and an increase in apoE-containing HDL, as related to the effect of LPS on a wide range of apolipoproteins, plasma enzymes, lipid transfer factors, and receptors that are involved in HDL metabolism. Reciprocally, all lipoprotein classes have been shown to bind LPS and to attenuate the biological response to LPS in vitro and in rodents. Moreover, triglyceride-rich lipoproteins protect rodents against death from LPS and bacterial sepsis. Accumulating evidence indicates that apolipoproteins such as apoE and apoAI, and not the lipid moieties of the particles, may be responsible for these protective effects of lipoproteins. Therefore, to increase our understanding of the complex interaction between lipoprotein metabolism and sepsis, further studies that address the specific roles of apolipoproteins in sepsis are warranted.
Collapse
Affiliation(s)
- Jimmy F.P. Berbee
- TNO-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands, Department of General Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Louis M. Havekes
- TNO-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C.N. Rensen
- TNO-Quality of Life, Department of Biomedical Research, Gaubius Laboratory, Leiden, The Netherlands, Department of General Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands,
| |
Collapse
|
38
|
Abstract
Animals that cannot sense endotoxin may die if they are infected by Gram-negative bacteria. Animals that sense endotoxin and respond too vigorously may also die, victims of their own inflammatory reactions. The outcome of Gram-negative bacterial infection is thus determined not only by an individual's ability to sense endotoxin and respond to its presence, but also by numerous phenomena that inactivate endotoxin and/or prevent harmful reactions to it. Endotoxin sensing requires the MD-2/TLR4 recognition complex and occurs principally in local tissues and the liver. This review highlights the known detoxification mechanisms, which include: (i) proteins that facilitate LPS sequestration by plasma lipoproteins, prevent interactions between the bioactive lipid A moiety and MD-2/TLR4, or promote cellular uptake via non-signaling pathway(s); (ii) enzymes that deacylate or dephosphorylate lipid A; (iii) mechanisms that remove LPS and Gram-negative bacteria from the bloodstream; and (iv) neuroendocrine adaptations that modulate LPS-induced mediator production or neutralize pro-inflammatory molecules in the circulation. In general, the mechanisms for sensing and detoxifying endotoxin seem to be compartmentalized (local versus systemic), dynamic, and variable between individuals. They may have evolved to confine infection and inflammation to extravascular sites of infection while preventing harmful systemic reactions. Integration of endotoxin sensing and detoxification is essential for successful host defense.
Collapse
Affiliation(s)
- Robert S. Munford
- Molecular Host Defense Laboratory, Departments of Internal Medicine and Microbiology, University of Texas Southwestern Medical School, Dallas, Texas, USA,
| |
Collapse
|
39
|
Thaveeratitham P, Plengpanich W, Naen-Udorn W, Patumraj S, Khovidhunkit W. Effects of human apolipoprotein A-I on endotoxin-induced leukocyte adhesion on endothelial cells in vivo and on the growth of Escherichia coli in vitro. ACTA ACUST UNITED AC 2016; 13:58-64. [PMID: 17621547 DOI: 10.1177/0968051907078611] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: High-density lipoprotein (HDL) has been shown to inhibit leukocyte adhesion to endothelial cells induced by endotoxin in vivo and suppress the growth of bacteria in vitro; however, the components responsible for these effects, either lipids or proteins, are not yet defined. In this study, we examined the effects of apolipoprotein (apo) A-I, the major protein of HDL, on ameliorating the effect of endotoxin and inhibiting the growth of bacteria. Materials and Methods: Apo A-I, purified from normal human HDL, was incubated with endotoxin. Leukocyte adhesion to endothelial cells of rat mesenteric venules was assessed using intravital fluorescence microscopy. Ability of apo A-I to inhibit the growth of Escherichia coli was assessed using a spread plate method. Results: Purified, lipid-free apo A-I could inhibit endotoxin-induced leukocyte adhesion to endothelial cells in vivo in a dose-dependent manner. In addition, apoA-I was able to suppress the growth of Escherichia coli in vitro. Conclusions: These data suggest that apo A-I of HDL can directly interact with endotoxin, ameliorating its effect and that apo A-I may have a direct toxic effect on whole bacteria. Therefore, therapeutic use of apo A-I in septicemia and bacterial infection should be further explored.
Collapse
Affiliation(s)
- Premtip Thaveeratitham
- Interdepartment of Physiology, Graduate School, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | | |
Collapse
|
40
|
Kitchens RL, Thompson PA, O'Keefe GE, Munford RS. Plasma constituents regulate LPS binding to, and release from, the monocyte cell surface. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519000060061101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Innate immunity to Gram-negative bacteria involves regulated mechanisms that allow sensitive but limited responses to LPS. Two important pathways that lead to host cell activation and LPS deactivation involve: (i) LPS interactions with CD14 and Toll-like receptor 4 on cells (activation); and (ii) LPS sequestration by plasma lipoproteins (deactivation). Whereas these pathways were previously thought to be independent and essentially irreversible, we found that they are connected by a third pathway: (iii) the movement of LPS from host cells to plasma lipoproteins. Our data show that, in the presence of human plasma, LPS binds transiently to monocyte surfaces and then moves from the cell surface to plasma lipoproteins. Soluble CD14 enhances LPS release from cells in the presence of lipoproteins, whereas LPS binding protein and phospholipid transfer protein do not. The transfer of cell-bound LPS to lipoproteins is accompanied by reduced cell responses to the LPS, suggesting that the movement of LPS from leukocytes into lipoproteins may attenuate host responses to LPS in vivo. Preliminary data suggest that changes that occur in the plasma after trauma or during sepsis decrease LPS binding to leukocytes while greatly increasing the rate of LPS release from cells.
Collapse
Affiliation(s)
- Richard L. Kitchens
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA,
| | - Patricia A. Thompson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Grant E. O'Keefe
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert S. Munford
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA, Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Azumi S, Tanamoto KI. Anti-endotoxin properties of a cinnamon bark-derived compound and its effect on the endotoxin shock model. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519990050030401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An endotoxin inhibitor derived from cinnamon bark was characterized chemically and tested for anti-LPS properties. Chemical analysis suggested that the active center of the inhibitor was in the lipid portion. Upon incubation with LPS molecule, the inhibitor reduced the ability of LPS to induce TNFα and generate nitric oxide from various cells in vitro, and Limulus gelation activity. The lethal toxicity of LPS in galactosamine-sensitized mice and pyrogenicity of LPS in a rabbit model were reduced 1000- and 100-fold by pre-incubation with the inhibitor, respectively. Simultaneous but separate injection of the inhibitor with a lethal dose of LPS also protected the majority of mice. Protection against LPS was seen when the inhibitor was given to mice 1 h before the LPS challenge. Furthermore, the inhibitor significantly suppressed the induction of fever by simultaneous administration with LPS without prior mixing. These results suggest that the inhibitor may be a useful potent blocker of bacterial endotoxin.
Collapse
Affiliation(s)
- Satoko Azumi
- Division of Microbiology, National Institute of Health Sciences, Tokyo, Japan
| | - Ken-ichi Tanamoto
- Division of Microbiology, National Institute of Health Sciences, Tokyo, Japan,
| |
Collapse
|
42
|
Harris HW, Gosnell JE, Kumwenda ZL. Review: The lipemia of sepsis: triglyceride-rich lipoproteins as agents of innate immunity. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519000060060301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Bacterial endotoxin (LPS) elicits dramatic responses in the host including elevated plasma lipid levels due to the increased synthesis and secretion of triglyceride (TG)-rich lipoproteins by the liver, and the inhibition of lipoprotein lipase. This cytokine-induced hyperlipoproteinemia, clinically termed the `lipemia of sepsis', was customarily thought to represent the mobilization of lipid stores to fuel the host response to infection. However, since lipoproteins can also bind and neutralize LPS, we hypothesize that TG-rich lipoproteins (VLDL and chylomicrons) are also components of an innate, non-adaptive host immune response to infection. Herein we review data demonstrating the capacity of lipoproteins to bind LPS, protect against LPS-induced toxicity, and modulate the overall host response to this bacterial toxin. Lastly, we propose a pathway whereby lipoprotein-bound LPS may represent a novel, endogenous mechanism for regulating the hepatic acute phase response.
Collapse
Affiliation(s)
- Hobart W. Harris
- UCSF Surgical Research Laboratory at San Francisco General Hospital, San Francisco, California, USA,
| | - Jessica E. Gosnell
- UCSF Surgical Research Laboratory at San Francisco General Hospital, San Francisco, California, USA, Department of Surgery, University of California, Davis-East Bay, Oakland, California, USA
| | - Zindaba L. Kumwenda
- UCSF Surgical Research Laboratory at San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|
43
|
Lemaire L, van der Poll T, van Lanschot J, Levels H, van der Ende B, van Deventer S, Gouma D. Human thoracic duct lymph inhibits lipopolysaccharide-induced release of cytokines. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199700400504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oral starvation causes gut atrophy and breakdown of barrier function, which can lead to transport of lipopolysaccharide (LPS) across the intestinal mucosa. Since triglyceride-rich lipoproteins can inhibit lipopolysaccharide-induced cytokine release, it can be hypothesized that enteral feeding protects the body against translocated LPS at the level of the thoracic duct by increasing the levels of triglycerides in thoracic duct lymph. We sought to determine the LPS-neutralizing capacity of human chyle by measuring LPS-induced cytokine production in vitro in the presence or absence of thoracic duct lymph. Moreover, we assessed whether enteral administration of triglyceride-rich lipoproteins changed the ability of lymph to influence LPS activity. Indeed, the presence of 10% and 100% triglyceride-poor and triglyceride-rich lymph induced a significant reduction in the TNF and IL-6 production elicited by LPS. However, there was no difference in the extent of inhibition of cytokine-release by triglyceride-poor and triglyceride-rich lymph. This study shows that lymph can inhibit LPS activity. This is not affected by prior enteral administration of triglycerides.
Collapse
Affiliation(s)
- L.C.J.M. Lemaire
- Department of Surgery, Laboratory of Experimental Internal Medicine
| | | | | | - H.M. Levels
- Center for Hemostasis, Thrombosis, Atherosclerosis and Inflammation Research, Academic Medical Center, Amsterdam, The Netherlands
| | - B. van der Ende
- Center for Hemostasis, Thrombosis, Atherosclerosis and Inflammation Research, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
44
|
Yao Z, Mates JM, Cheplowitz AM, Hammer LP, Maiseyeu A, Phillips GS, Wewers MD, Rajaram MVS, Robinson JM, Anderson CL, Ganesan LP. Blood-Borne Lipopolysaccharide Is Rapidly Eliminated by Liver Sinusoidal Endothelial Cells via High-Density Lipoprotein. THE JOURNAL OF IMMUNOLOGY 2016; 197:2390-9. [PMID: 27534554 DOI: 10.4049/jimmunol.1600702] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/14/2016] [Indexed: 12/12/2022]
Abstract
During Gram-negative bacterial infections, excessive LPS induces inflammation and sepsis via action on immune cells. However, the bulk of LPS can be cleared from circulation by the liver. Liver clearance is thought to be a slow process mediated exclusively by phagocytic resident macrophages, Kupffer cells (KC). However, we discovered that LPS disappears rapidly from the circulation, with a half-life of 2-4 min in mice, and liver eliminates about three quarters of LPS from blood circulation. Using microscopic techniques, we found that ∼75% of fluor-tagged LPS in liver became associated with liver sinusoidal endothelial cells (LSEC) and only ∼25% with KC. Notably, the ratio of LSEC-KC-associated LPS remained unchanged 45 min after infusion, indicating that LSEC independently processes the LPS. Most interestingly, results of kinetic analysis of LPS bioactivity, using modified limulus amebocyte lysate assay, suggest that recombinant factor C, an LPS binding protein, competitively inhibits high-density lipoprotein (HDL)-mediated LPS association with LSEC early in the process. Supporting the previous notion, 3 min postinfusion, 75% of infused fluorescently tagged LPS-HDL complex associates with LSEC, suggesting that HDL facilitates LPS clearance. These results lead us to propose a new paradigm of LSEC and HDL in clearing LPS with a potential to avoid inflammation during sepsis.
Collapse
Affiliation(s)
- Zhili Yao
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Jessica M Mates
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Alana M Cheplowitz
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Lindsay P Hammer
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Andrei Maiseyeu
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, MD 21201
| | - Gary S Phillips
- Department of Biostatistics, The Ohio State University, Columbus, OH 43210
| | - Mark D Wewers
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Murugesan V S Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
| | - John M Robinson
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210
| | - Clark L Anderson
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Latha P Ganesan
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210;
| |
Collapse
|
45
|
Foit L, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles potently inhibit cell signaling and production of inflammatory mediators induced by lipopolysaccharide binding Toll-like receptor 4. Biomaterials 2016; 100:67-75. [PMID: 27244690 DOI: 10.1016/j.biomaterials.2016.05.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/25/2022]
Abstract
Toll-like receptor 4 (TLR4) plays a critical role in the innate immune system. Stimulation of TLR4 occurs upon binding lipopolysaccharide (LPS), a component of Gram-negative bacterial cell walls. Due to the potency of the induced inflammatory response, there is a growing interest in agents that can most proximally modulate this LPS/TLR4 interaction to prevent downstream cell signaling events and the production of inflammatory mediators. Building on the natural ability of human high-density lipoprotein (HDL) to bind LPS, we synthesized a suite of HDL-like nanoparticles (HDL-like NP). We identified one HDL-like NP that was particularly effective at decreasing TLR4 signaling caused by addition of purified LPS or Gram-negative bacteria to model human cell lines or primary human peripheral blood cells. The HDL-like NP functioned to inhibit TLR4-dependent inflammatory response to LPS derived from multiple bacterial species. Mechanistically, data show that the NP mainly functions by scavenging and neutralizing the LPS toxin. Taken together, HDL-like NPs constitute a powerful endotoxin scavenger with the potential to significantly reduce LPS-mediated inflammation.
Collapse
Affiliation(s)
- Linda Foit
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA
| | - C Shad Thaxton
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA; International Institute for Nanotechnology (IIN), 2145 Sheridan Road, Evanston, IL 60208, USA; Robert H Lurie Comprehensive Cancer Center (RHLCCC), Northwestern University, Feinberg School of Medicine, 303 E Superior, Chicago, IL 60611, USA.
| |
Collapse
|
46
|
Zhou H, Hu J, Zhu Q, Yang S, Zhang Y, Gao R, Liu L, Wang Y, Zhen Q, Lv Q, Li Q. Lipopolysaccharide-binding protein cannot independently predict type 2 diabetes mellitus: A nested case-control study. J Diabetes 2016; 8:214-9. [PMID: 25753130 DOI: 10.1111/1753-0407.12281] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/30/2015] [Accepted: 02/20/2015] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Cross-sectional studies have reported a close association between serum lipopolysaccharide-binding protein (LBP) and many metabolic disorders. However, no longitudinal study has explored the relationship between LBP and type 2 diabetes mellitus (T2DM). The aim of the present study was to investigate the association between serum LBP levels and the risk of developing T2DM. METHODS A 5-year nested case-control study of 3510 individuals was performed as part of the Environment, Inflammation and Metabolic Diseases Study (EIMDS). Clinical data were collected at baseline. Serum levels of LBP and other biochemical factors, such as insulin and high-sensitivity C-reactive protein, were detected 5 years later. Subjects were diagnosed as having T2DM on the basis of results of an oral glucose tolerance test (OGTT) and 1998 World Health Organization criteria. Controls were randomly selected to match cases according to age, gender, and body mass index (BMI) in a 1:1 ratio. Odds ratios (OR) for T2DM were calculated using conditional logistic regression analysis. RESULTS Over a 5-year follow-up period, 255 subjects developed T2DM. There was no significant difference in serum LBP levels between the T2DM and control groups at baseline (19.78 ± 6.40 versus 20.53 ± 6.99 μg/mL; P = 0.207). Subjects were divided into three groups based on tertiles of LBP concentrations (n = 170 in each group; T1 = 1.31-17.16 μg/mL LBP; T2 = 17.21-22.37 μg/mL LBP; T3 = 22.49-40.08 μg/mL LBP). There was no significant association between the risk of developing T2DM and any of the LBP tertiles in either a simple model or after adjusting for general status and biochemical factors. CONCLUSION After matching for gender, age, and BMI, LBP does not improve prediction of the development of T2DM independently.
Collapse
Affiliation(s)
- Huang Zhou
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinbo Hu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qibo Zhu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shumin Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Zhang
- Hospital of Chongqing University, Chongqing, China
| | - Rufei Gao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lulu Liu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianna Zhen
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiong Lv
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qifu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
47
|
Impact of prebiotic supplementation on T-cell subsets and their related cytokines, anthropometric features and blood pressure in patients with type 2 diabetes mellitus: A randomized placebo-controlled Trial. Complement Ther Med 2016; 24:96-102. [DOI: 10.1016/j.ctim.2015.12.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 12/12/2015] [Accepted: 12/21/2015] [Indexed: 01/27/2023] Open
|
48
|
Gnauck A, Lentle RG, Kruger MC. Chasing a ghost?--Issues with the determination of circulating levels of endotoxin in human blood. Crit Rev Clin Lab Sci 2016; 53:197-215. [PMID: 26732012 DOI: 10.3109/10408363.2015.1123215] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reliable quantification of bacterial products such as endotoxin is important for the diagnosis of Gram-negative infection and for the monitoring of its treatment. Further, it is important to identify patients with persistent subclinical level of bacterial products in their systemic circulation as data from animal studies also suggest this may be correlated with the onset of metabolic syndrome. In this review, we first aim to describe the principles of the Limulus amoebocyte lysate (LAL) test, an assay that is used to quantify endotoxin, and the various shortcomings that must be addressed before it can become a reliable means of quantifying endotoxin in samples derived from blood. We then review published data regarding endotoxin levels in healthy subjects and those with sepsis, inflammatory bowel disease, liver disorders and metabolic disorders such as obesity and diabetes. We also review the evidence regarding influence of macronutrients in augmenting the levels of systemic endotoxin. The results of this review show that reported mean levels of endotoxin in the systemic circulation of healthy humans and of those with various clinical disorders vary over a wide range. Further, this review shows that a significant proportion of this variation can be related to the method that was used to prepare plasma and serum samples prior to assay and its ability to reduce the effect of various blood borne factors that interfere with the LAL assay.
Collapse
Affiliation(s)
- Anne Gnauck
- a Physiology Group, School of Food and Nutrition, College of Health, Massey University , Palmerston North , New Zealand
| | - Roger Graham Lentle
- a Physiology Group, School of Food and Nutrition, College of Health, Massey University , Palmerston North , New Zealand
| | - Marlena Cathorina Kruger
- a Physiology Group, School of Food and Nutrition, College of Health, Massey University , Palmerston North , New Zealand
| |
Collapse
|
49
|
Constantinou C, Karavia EA, Xepapadaki E, Petropoulou PI, Papakosta E, Karavyraki M, Zvintzou E, Theodoropoulos V, Filou S, Hatziri A, Kalogeropoulou C, Panayiotakopoulos G, Kypreos KE. Advances in high-density lipoprotein physiology: surprises, overturns, and promises. Am J Physiol Endocrinol Metab 2016; 310:E1-E14. [PMID: 26530157 DOI: 10.1152/ajpendo.00429.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/30/2015] [Indexed: 12/21/2022]
Abstract
Emerging evidence strongly supports that changes in the HDL metabolic pathway, which result in changes in HDL proteome and function, appear to have a causative impact on a number of metabolic disorders. Here, we provide a critical review of the most recent and novel findings correlating HDL properties and functionality with various pathophysiological processes and disease states, such as obesity, type 2 diabetes mellitus, nonalcoholic fatty liver disease, inflammation and sepsis, bone and obstructive pulmonary diseases, and brain disorders.
Collapse
Affiliation(s)
| | - Eleni A Karavia
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Eva Xepapadaki
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | | | - Eugenia Papakosta
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Marilena Karavyraki
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Evangelia Zvintzou
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | | | - Serafoula Filou
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Aikaterini Hatziri
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | | | | | - Kyriakos E Kypreos
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| |
Collapse
|
50
|
Gnauck A, Lentle RG, Kruger MC. The Characteristics and Function of Bacterial Lipopolysaccharides and Their Endotoxic Potential in Humans. Int Rev Immunol 2015; 35:189-218. [PMID: 26606737 DOI: 10.3109/08830185.2015.1087518] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cross-talk between enteral microbiota and human host is essential for the development and maintenance of the human gastrointestinal and systemic immune systems. The presence of lipopolysaccharides (LPS) lysed from the cell membrane of Gram-negative bacteria in the gut lumen is thought to promote the development of a balanced gut immune response whilst the entry of the same LPS into systemic circulation may lead to a deleterious pro-inflammatory systemic immune response. Recent data suggest that chronically low levels of circulating LPS may be associated with the development of metabolic diseases such as insulin resistance, type 2 diabetes, atherosclerosis and cardiovascular disease. This review focuses on the cross-talk between enteral commensal bacteria and the human immune system via LPS. We explain the structural characterisation of the LPS molecule and its function in the bacteria. We then examine how LPS is recognised by various elements of the human immune system and the signalling pathways that are activated by the structure of the LPS molecule and the effect of various concentrations. Further, we discuss the sequelae of this signalling in the gut-associated and systemic immune systems i.e. the neutralisation of LPS and the development of tolerance to LPS.
Collapse
Affiliation(s)
- Anne Gnauck
- a School of Food and Nutrition, College of Health , Massey University , Palmerston North , New Zealand
| | - Roger G Lentle
- a School of Food and Nutrition, College of Health , Massey University , Palmerston North , New Zealand
| | - Marlena C Kruger
- a School of Food and Nutrition, College of Health , Massey University , Palmerston North , New Zealand
| |
Collapse
|