1
|
Escudero B, López-Valencia L, Arias Horcajadas F, Orio L. Divergent Roles of APOAI and APOM in the Identification of Alcohol Use Disorder and Their Association With Inflammation and Cognitive Decline: A Pilot Study. Int J Neuropsychopharmacol 2024; 27:pyae029. [PMID: 38970624 PMCID: PMC11287869 DOI: 10.1093/ijnp/pyae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Alcohol use disorder (AUD) courses with inflammation and cognitive decline. Apolipoproteins have emerged as novel target compounds related to inflammatory processes and cognition. METHODS A cross-sectional study was performed on abstinent AUD patients with at least 1 month of abstinence (n = 33; 72.7% men) and healthy controls (n = 34; 47.1% men). A battery of plasma apolipoproteins (APOAI, APOAII, APOB, APOCII, APOE, APOJ, and APOM), plasma inflammatory markers (LPS, LBP), and their influence on cognition and presence of the disorder were investigated. RESULTS Higher levels of plasma APOAI, APOB, APOE, and APOJ, as well as the proinflammatory LPS, were observed in the AUD group, irrespective of sex, whereas APOM levels were lower vs controls. Hierarchical logistic regression analyses, adjusting for covariates (age, sex, education), associated APOM with the absence of cognitive impairment in AUD and identified APOAI and APOM as strong predictors of the presence or absence of the disorder, respectively. APOAI and APOM did not correlate with alcohol abuse variables or liver status markers, but they showed an opposite profile in their associations with LPS (positive for APOAI; negative for APOM) and cognition (negative for APOAI; positive for APOM) in the entire sample. CONCLUSIONS The HDL constituents APOAI and APOM were differentially regulated in the plasma of AUD patients compared with controls, playing divergent roles in the disorder identification and associations with inflammation and cognitive decline.
Collapse
Affiliation(s)
- Berta Escudero
- Instituto de investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain
| | - Leticia López-Valencia
- Instituto de investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain
| | - Francisco Arias Horcajadas
- Instituto de investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Riapad: Research Network in Primary Care in Addictions, Spain
| | - Laura Orio
- Instituto de investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain
- Riapad: Research Network in Primary Care in Addictions, Spain
| |
Collapse
|
2
|
Zhang X, van der Vorst EPC. High-Density Lipoprotein Modifications: Causes and Functional Consequences in Type 2 Diabetes Mellitus. Cells 2024; 13:1113. [PMID: 38994965 PMCID: PMC11240616 DOI: 10.3390/cells13131113] [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: 05/31/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
High-density lipoprotein (HDL) is a group of small, dense, and protein-rich lipoproteins that play a role in cholesterol metabolism and various cellular processes. Decreased levels of HDL and HDL dysfunction are commonly observed in individuals with type 2 diabetes mellitus (T2DM), which is also associated with an increased risk for cardiovascular disease (CVD). Due to hyperglycemia, oxidative stress, and inflammation that develop in T2DM, HDL undergoes several post-translational modifications such as glycation, oxidation, and carbamylation, as well as other alterations in its lipid and protein composition. It is increasingly recognized that the generation of HDL modifications in T2DM seems to be the main cause of HDL dysfunction and may in turn influence the development and progression of T2DM and its related cardiovascular complications. This review provides a general introduction to HDL structure and function and summarizes the main modifications of HDL that occur in T2DM. Furthermore, the potential impact of HDL modifications on the pathogenesis of T2DM and CVD, based on the altered interactions between modified HDL and various cell types that are involved in glucose homeostasis and atherosclerotic plaque generation, will be discussed. In addition, some perspectives for future research regarding the T2DM-related HDL modifications are addressed.
Collapse
Affiliation(s)
- Xiaodi Zhang
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), 80336 Munich, Germany
| |
Collapse
|
3
|
Carmo HRP, Bonilha I, Barreto J, Tognolini M, Zanotti I, Sposito AC. High-Density Lipoproteins at the Interface between the NLRP3 Inflammasome and Myocardial Infarction. Int J Mol Sci 2024; 25:1290. [PMID: 38279290 PMCID: PMC10816227 DOI: 10.3390/ijms25021290] [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: 12/30/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Despite significant therapeutic advancements, morbidity and mortality following myocardial infarction (MI) remain unacceptably high. This clinical challenge is primarily attributed to two significant factors: delayed reperfusion and the myocardial injury resulting from coronary reperfusion. Following reperfusion, there is a rapid intracellular pH shift, disruption of ionic balance, heightened oxidative stress, increased activity of proteolytic enzymes, initiation of inflammatory responses, and activation of several cell death pathways, encompassing apoptosis, necroptosis, and pyroptosis. The inflammatory cell death or pyroptosis encompasses the activation of the intracellular multiprotein complex known as the NLRP3 inflammasome. High-density lipoproteins (HDL) are endogenous particles whose components can either promote or mitigate the activation of the NLRP3 inflammasome. In this comprehensive review, we explore the role of inflammasome activation in the context of MI and provide a detailed analysis of how HDL can modulate this process.
Collapse
Affiliation(s)
- Helison R. P. Carmo
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Isabella Bonilha
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Joaquim Barreto
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | | | - Ilaria Zanotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Andrei C. Sposito
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| |
Collapse
|
4
|
Muendlein A, Heinzle C, Brandtner EM, Leiherer A, Geiger K, Gaenger S, Drexel H, Dechow T, Decker T. Plasma apolipoprotein M predicts overall survival in metastatic breast cancer patients. Breast Cancer Res Treat 2023; 201:571-576. [PMID: 37490173 DOI: 10.1007/s10549-023-07045-4] [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: 05/10/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Apolipoprotein M (APOM) is a plasma apolipoprotein closely involved with lipid metabolism and inflammation. In vitro studies suggest that APOM may also have a tumor-suppressive role in breast cancer. In the present study, we aimed to evaluate the impact of plasma APOM levels on the prognosis of breast cancer patients. METHODS We measured APOM levels using an enzyme-linked immunosorbent assay in 75 patients with ER-positive/HER2-negative metastatic breast cancer. The endpoint was overall survival (OS) at 24 months. RESULTS During the 24-month follow-up period, 34.7% of the patients died. Baseline APOM levels were significantly reduced in patients who deceased during follow-up compared to survivors (42.7 ± 14.5 µg/mL versus 52.2 ± 13.8 µg/mL; P = 0.003). Cox regression analysis showed a hazard ratio of 0.30 [95% confidence interval 0.15-0.61]; P < 0.001 per doubling of APOM levels. Correction for age, C-reactive protein, menopausal state, histology of the primary tumor, metastatic site, number of metastases, endocrine resistance, scheduled therapy line, and kind of scheduled therapy indicated that circulating APOM predicted OS independently of these parameters (HRper doubling = 0.23 [0.09-0.56; P = 0.001). CONCLUSIONS Our study suggests that circulating APOM is significantly linked with reduced mortality in metastatic breast cancer patients.
Collapse
Affiliation(s)
- Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria.
| | - Christine Heinzle
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria
- Medical Central Laboratories, Feldkirch, Austria
| | - Eva Maria Brandtner
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria
- Medical Central Laboratories, Feldkirch, Austria
| | - Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria
- Medical Central Laboratories, Feldkirch, Austria
| | - Stella Gaenger
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment Laboratory, Stadtstrasse 33, 6850, Dornbirn, Austria
- Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | |
Collapse
|
5
|
Drexler Y, Molina J, Elfassy T, Ma R, Christoffersen C, Kurano M, Yatomi Y, Mariani LH, Contreras G, Merscher S, Fornoni A. Identification of Glomerular and Plasma Apolipoprotein M as Novel Biomarkers in Glomerular Disease. Kidney Int Rep 2023; 8:884-897. [PMID: 37069998 PMCID: PMC10105063 DOI: 10.1016/j.ekir.2023.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Dysregulation of sphingolipid and cholesterol metabolism contributes to the pathogenesis of glomerular diseases (GDs). Apolipoprotein M (ApoM) promotes cholesterol efflux and modulates the bioactive sphingolipid sphingosine-1-phosphate (S1P). Glomerular ApoM expression is decreased in patients with focal segmental glomerulosclerosis (FSGS). We hypothesized that glomerular ApoM deficiency occurs in GD and that ApoM expression and plasma ApoM correlate with outcomes. Methods Patients with GD from the Nephrotic Syndrome Study Network (NEPTUNE) were studied. We compared glomerular mRNA expression of ApoM (gApoM), sphingosine kinase 1 (SPHK1), and S1P receptors 1 to 5 (S1PR1-5) in patients (n = 84) and controls (n = 6). We used correlation analyses to determine associations between gApoM, baseline plasma ApoM (pApoM), and urine ApoM (uApoM/Cr). We used linear regression to determine whether gApoM, pApoM, and uApoM/Cr were associated with baseline estimated glomerular filtration rate (eGFR) and proteinuria. Using Cox models, we determined whether gApoM, pApoM, and uApoM/Cr were associated with complete remission (CR) and the composite of end-stage kidney disease (ESKD) or ≥40% eGFR decline. Results gApoM was reduced (P < 0.01) and SPHK1 and S1PR1 to 5 expression was increased (P < 0.05) in patients versus controls, consistent with ApoM/S1P pathway modulation. gApoM positively correlated with pApoM in the overall cohort (r = 0.34, P < 0.01) and in the FSGS (r = 0.48, P < 0.05) and minimal change disease (MCD) (r = 0.75, P < 0.05) subgroups. Every unit decrease in gApoM and pApoM (log2) was associated with a 9.77 ml/min per 1.73 m2 (95% confidence interval [CI]: 3.96-15.57) and 13.26 ml/min per 1.73 m2 (95% CI: 3.57-22.96) lower baseline eGFR, respectively (P < 0.01). From Cox models adjusted for age, sex, or race, pApoM was a significant predictor of CR (hazard ratio [HR]: 1.85; 95% CI: 1.06-3.23). Conclusions pApoM is a potential noninvasive biomarker of gApoM deficiency and strongly associates with clinical outcomes in GD.
Collapse
Affiliation(s)
- Yelena Drexler
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Tali Elfassy
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ruixuan Ma
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Christina Christoffersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Laura H. Mariani
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Gabriel Contreras
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
6
|
Association of apolipoprotein M and sphingosine-1-phosphate with brown adipose tissue after cold exposure in humans. Sci Rep 2022; 12:18753. [PMID: 36335116 PMCID: PMC9637161 DOI: 10.1038/s41598-022-21938-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
The HDL-associated apolipoprotein M (apoM) and its ligand sphingosine-1-phosphate (S1P) may control energy metabolism. ApoM deficiency in mice is associated with increased vascular permeability, brown adipose tissue (BAT) mass and activity, and protection against obesity. In the current study, we explored the connection between plasma apoM/S1P levels and parameters of BAT as measured via 18F-FDG PET/CT after cold exposure in humans. Fixed (n = 15) vs personalized (n = 20) short-term cooling protocols decreased and increased apoM (- 8.4%, P = 0.032 vs 15.7%, P < 0.0005) and S1P (- 41.0%, P < 0.0005 vs 19.1%, P < 0.005) plasma levels, respectively. Long-term cooling (n = 44) did not affect plasma apoM or S1P levels. Plasma apoM and S1P did not correlate significantly to BAT volume and activity in the individual studies. However, short-term studies combined, showed that increased changes in plasma apoM correlated with BAT metabolic activity (β: 0.44, 95% CI [0.06-0.81], P = 0.024) after adjusting for study design but not BAT volume (β: 0.39, 95% CI [- 0.01-0.78], P = 0.054). In conclusion, plasma apoM and S1P levels are altered in response to cold exposure and may be linked to changes in BAT metabolic activity but not BAT volume in humans. This contrasts partly with observations in animals and highlights the need for further studies to understand the biological role of apoM/S1P complex in human adipose tissue and lipid metabolism.
Collapse
|
7
|
Sphingosine 1-phosphate receptor-targeted therapeutics in rheumatic diseases. Nat Rev Rheumatol 2022; 18:335-351. [PMID: 35508810 DOI: 10.1038/s41584-022-00784-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphate (S1P), which acts via G protein-coupled S1P receptors (S1PRs), is a bioactive lipid essential for vascular integrity and lymphocyte trafficking. The S1P-S1PR signalling axis is a key component of the inflammatory response in autoimmune rheumatic diseases. Several drugs that target S1PRs have been approved for the treatment of multiple sclerosis and inflammatory bowel disease and are under clinical testing for patients with systemic lupus erythematosus (SLE). Preclinical studies support the hypothesis that targeting the S1P-S1PR axis would be beneficial to patients with SLE, rheumatoid arthritis (RA) and systemic sclerosis (SSc) by reducing pathological inflammation. Whereas most preclinical research and development efforts are focused on reducing lymphocyte trafficking, protective effects of circulating S1P on endothelial S1PRs, which maintain the vascular barrier and enable blood circulation while dampening leukocyte extravasation, have been largely overlooked. In this Review, we take a holistic view of S1P-S1PR signalling in lymphocyte and vascular pathobiology. We focus on the potential of S1PR modulators for the treatment of SLE, RA and SSc and summarize the rationale, pathobiology and evidence from preclinical models and clinical studies. Improved understanding of S1P pathobiology in autoimmune rheumatic diseases and S1PR therapeutic modulation is anticipated to lead to efficacious and safer management of these diseases.
Collapse
|
8
|
Diab A, Valenzuela Ripoll C, Guo Z, Javaheri A. HDL Composition, Heart Failure, and Its Comorbidities. Front Cardiovasc Med 2022; 9:846990. [PMID: 35350538 PMCID: PMC8958020 DOI: 10.3389/fcvm.2022.846990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Although research on high-density lipoprotein (HDL) has historically focused on atherosclerotic coronary disease, there exists untapped potential of HDL biology for the treatment of heart failure. Anti-oxidant, anti-inflammatory, and endothelial protective properties of HDL could impact heart failure pathogenesis. HDL-associated proteins such as apolipoprotein A-I and M may have significant therapeutic effects on the myocardium, in part by modulating signal transduction pathways and sphingosine-1-phosphate biology. Furthermore, because heart failure is a complex syndrome characterized by multiple comorbidities, there are complex interactions between heart failure, its comorbidities, and lipoprotein homeostatic mechanisms. In this review, we will discuss the effects of heart failure and associated comorbidities on HDL, explore potential cardioprotective properties of HDL, and review novel HDL therapeutic targets in heart failure.
Collapse
|
9
|
Zou G, Zhu Q, Ren B, Guo Q, Wu Y, He J, Wu Y, Luo Z. HDL-Associated Lipoproteins: Potential Prognostic Biomarkers for Gram-Negative Sepsis. J Inflamm Res 2022; 15:1117-1131. [PMID: 35210815 PMCID: PMC8860992 DOI: 10.2147/jir.s350737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/28/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose To determine the levels of serum HDL-associated apolipoproteins (apoM and apoC) and HDL-binding receptor (scavenger receptor BI, SR-BI) in patients with gram-negative bacteria sepsis (G-sepsis) and to evaluate the value of lipoproteins in the diagnosis, severity and prognosis of G-sepsis. Patients and Methods A total of 128 patients with sepsis, 40 patients with system inflammatory reaction syndrome (SIRS) and 40 healthy subjects were enrolled in the Second People’s Hospital of Hunan Province from September 2019 to September 2020. The levels and the correlation of lipoproteins were detected and dynamically monitored by enzyme-linked adsorption method, ROC curve for the diagnostic, severity and prognostic value of lipoproteins in G-sepsis. Results The levels of serum HDL-associated lipoproteins in patients with G-sepsis were significantly decreased (P < 0.05), and the ROC curve showed that HDL-C, SR-BI, apoM and apoC had cut-off values of 0.915 mmol/L, 122.100 pg/mL, 102.400 ug/mL and 17.55 mg/mL, respectively, for the diagnosis of G-sepsis, with the sensitivity was 85.56%, 97.78%, 93.33% and 73.03%, and the specificity was 95.0%, 82.50%, 61.54% and 82.50%, respectively. There was a correlation between HDL-associated apolipoproteins. Changes in serum HDL-associated lipoproteins were more obvious in shock group than classic inflammation indicators, such as PCT, IL-6 and CRP. They showed a trend change on day 3, with the levels of SR-BI and apoC changing 2–3 times, and the sensitivity of HDL-C, SR-BI, apoM and apoC for the diagnosis of G-septic shock were 32.43%, 72.97%, 65.75%, and 43.24%, and specificity of 94.34%, 81.13%, 83.07%, and 86.79%, respectively. The AUC, sensitivity and specificity of apoM combined with SR-BI were improved. Conclusion HDL-associated lipoproteins were correlated with bacterial-infected types, and serum levels of HDL-associated lipoproteins can be used as potential biomarkers for early diagnosis and progress of G-sepsis. ApoM combined with SR-BI could improve the sensitivity and specificity of prognosis assessment.
Collapse
Affiliation(s)
- Guoying Zou
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Qing Zhu
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Biqiong Ren
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Qi Guo
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Yuanyuan Wu
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Junyu He
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Ying Wu
- Department of Clinical Laboratory, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
| | - Zhihong Luo
- Office of the Party Committee, The Second People’s Hospital of Hunan Province, Changsha, Hunan, 410007, People’s Republic of China
- Correspondence: Zhihong Luo, Office of the Party Committee, The Second People’s Hospital of Hunan Province, Furong Middle Road 427, Yuhua District, Changsha, Hunan, 410007, People’s Republic of China, Tel +86 19848029533, Email
| |
Collapse
|
10
|
Bisgaard LS, Christoffersen C. The apoM/S1P Complex-A Mediator in Kidney Biology and Disease? Front Med (Lausanne) 2021; 8:754490. [PMID: 34722589 PMCID: PMC8553247 DOI: 10.3389/fmed.2021.754490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Kidney disease affects more than 10% of the population, can be both acute and chronic, and is linked to other diseases such as cardiovascular disease, diabetes, and sepsis. Despite the detrimental consequences for patients, no good treatment options directly targeting the kidney are available. Thus, a better understanding of the pathology and new treatment modalities are required. Accumulating evidence suggests that the apolipoprotein M/sphingosine-1-phosphate (apoM/S1P) axis is a likely drug target, but significant gaps in our knowledge remain. In this review, we present what has so far been elucidated about the role of apoM in normal kidney biology and describe how changes in the apoM/S1P axis are thought to affect the development of kidney disease. ApoM is primarily produced in the liver and kidneys. From the liver, apoM is secreted into circulation, where it is attached to lipoproteins (primarily HDL). Importantly, apoM is a carrier of the bioactive lipid S1P. S1P acts by binding to five different receptors. Together, apoM/S1P plays a role in several biological mechanisms, such as inflammation, endothelial cell permeability, and lipid turnover. In the kidney, apoM is primarily expressed in the proximal tubular cells. S1P can be produced locally in the kidney, and several of the five S1P receptors are present in the kidney. The functional role of kidney-derived apoM as well as plasma-derived apoM is far from elucidated and will be discussed based on both experimental and clinical studies. In summary, the current studies provide evidence that support a role for the apoM/S1P axis in kidney disease; however, additional pre-clinical and clinical studies are needed to reveal the mechanisms and target potential in the treatment of patients.
Collapse
Affiliation(s)
- Line S Bisgaard
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Su X, Zhang G, Cheng Y, Wang B. New insights into the emerging effects of inflammatory response on HDL particles structure and function. Mol Biol Rep 2021; 48:5723-5733. [PMID: 34319542 DOI: 10.1007/s11033-021-06553-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
According to the increasing results, it has been well-demonstrated that the chronic inflammatory response, including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism whereby inflammatory response up-regulates the risk of cardio-metabolic disorder disease is multifactorial; furthermore, the alterations in high density lipoprotein (HDL) structure and function which occur under the inflammatory response could play an important modulatory function. On the other hand, the serum concentrations of HDL cholesterol (HDL-C) have been shown to be reduced significantly under inflammatory status with remarked alterations in HDL particles. Nevertheless, the potential mechanism whereby the inflammatory response reduces serum HDL-C levels is not simply defined but reduces apolipoprotein A1 production. The alterations in HDL structure mediated by the inflammatory response has been also confirmed to decrease the ability of HDL particle to play an important role in reverse cholesterol transport and protect the LDL particles from oxidation. Recently, it has been shown that under the inflammatory condition, diverse alterations in HDL structure could be observed which lead to changes in HDL function. In the current review, the emerging effects of inflammatory response on HDL particles structure and function are well-summarized to elucidate the potential mechanism whereby different inflammatory status modulates the pathogenic development of dyslipidemia.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Guoming Zhang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Ye Cheng
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| | - Bin Wang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| |
Collapse
|
12
|
Karlsson MJ, Costa Svedman F, Tebani A, Kotol D, Höiom V, Fagerberg L, Edfors F, Uhlén M, Egyhazi Brage S, Maddalo G. Inflammation and Apolipoproteins Are Potential Biomarkers for Stratification of Cutaneous Melanoma Patients for Immunotherapy and Targeted Therapy. Cancer Res 2021; 81:2545-2555. [PMID: 33574091 DOI: 10.1158/0008-5472.can-20-2000] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/22/2020] [Accepted: 02/09/2021] [Indexed: 11/16/2022]
Abstract
Malignant cutaneous melanoma is one of the most common cancers in young adults. During the last decade, targeted and immunotherapies have significantly increased the overall survival of patients with malignant cutaneous melanoma. Nevertheless, disease progression is common, and a lack of predictive biomarkers of patient response to therapy hinders individualized treatment strategies. To address this issue, we performed a longitudinal study using an unbiased proteomics approach to identify and quantify proteins in plasma both before and during treatment from 109 patients treated with either targeted or immunotherapy. Linear modeling and machine learning approaches identified 43 potential prognostic and predictive biomarkers. A reverse correlation between apolipoproteins and proteins related to inflammation was observed. In the immunotherapy group, patients with low pretreatment expression of apolipoproteins and high expression of inflammation markers had shorter progression-free survival. Similarly, increased expression of LDHB during treatment elicited a significant impact on response to immunotherapy. Overall, we identified potential common and treatment-specific biomarkers in malignant cutaneous melanoma, paving the way for clinical use of these biomarkers following validation on a larger cohort. SIGNIFICANCE: This study identifies a potential biomarker panel that could improve the selection of therapy for patients with cutaneous melanoma.
Collapse
Affiliation(s)
- Max J Karlsson
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Abdellah Tebani
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - David Kotol
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Gianluca Maddalo
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
13
|
Mondal P, Aljizeeri A, Small G, Malhotra S, Harikrishnan P, Affandi JS, Buechel RR, Dwivedi G, Al-Mallah MH, Jain D. Coronary artery disease in patients with human immunodeficiency virus infection. J Nucl Cardiol 2021; 28:510-530. [PMID: 32820424 DOI: 10.1007/s12350-020-02280-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/01/2020] [Indexed: 01/02/2023]
Abstract
The life expectancy of people infected with human immunodeficiency virus (HIV) is rising due to better access to combination anti-retroviral therapy (ART). Although ART has reduced acquired immune deficiency syndrome (AIDS) related mortality and morbidity, there has been an increase in non-AIDS defining illnesses such as diabetes mellitus, hypercholesterolemia and coronary artery disease (CAD). HIV is a disease marked by inflammation which has been associated with specific biological vascular processes increasing the risk of premature atherosclerosis. The combination of pre-existing risk factors, atherosclerosis, ART, opportunistic infections and coagulopathy contributes to rising CAD incidence. The prevalence of CAD has emerged as a major contributor of morbidity in these patients due to longer life expectancy. However, ART has been associated with lipodystrophy, dyslipidemia, insulin resistance, diabetes mellitus and CAD. These adverse effects, along with drug-drug interactions when ART is combined with cardiovascular drugs, result in significant challenges in the care of this group of patients. Exercise tolerance testing, echocardiography, myocardial perfusion imaging, coronary computed tomography angiography and magnetic resonance imaging help in the diagnosis of CAD and heart failure and help predict cardiovascular outcomes in a manner similar to non-infected individuals. This review will highlight the pathogenesis and factors that link HIV to CAD, presentation and treatment of HIV-patients presenting with CAD and review briefly the cardiac imaging modalities used to identify this entity and help prognosticate future outcomes.
Collapse
Affiliation(s)
- Pratik Mondal
- Department of Cardiology and Nuclear Cardiovascular Imaging Laboratory, New York Medical College, Westchester Medical Center, 100 Woods Road, Valhalla, NY, 10595, USA
| | - Ahmed Aljizeeri
- King Abdulaziz Cardiac Center, Ministry of National Guard-Health Affaire, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Gary Small
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Saurabh Malhotra
- Division of Cardiology, Cook County Health, Chicago, IL, USA
- Division of Cardiology, Rush Medical College, Chicago, IL, USA
| | | | | | - Ronny R Buechel
- Department of Nuclear Medicine, Cardiac Imaging, University Hospital Zurich, Zurich, Switzerland
| | - Girish Dwivedi
- Fiona Stanley Hospital, Murdoch, WA, Australia
- Harry Perkins Institute of Medical Research, Murdoch, WA, Australia
- The University of Western Australia, Crawley, WA, Australia
| | - Mouaz H Al-Mallah
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, USA
| | - Diwakar Jain
- Department of Cardiology and Nuclear Cardiovascular Imaging Laboratory, New York Medical College, Westchester Medical Center, 100 Woods Road, Valhalla, NY, 10595, USA.
| |
Collapse
|
14
|
Morris G, Puri BK, Bortolasci CC, Carvalho A, Berk M, Walder K, Moreira EG, Maes M. The role of high-density lipoprotein cholesterol, apolipoprotein A and paraoxonase-1 in the pathophysiology of neuroprogressive disorders. Neurosci Biobehav Rev 2021; 125:244-263. [PMID: 33657433 DOI: 10.1016/j.neubiorev.2021.02.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Lowered high-density lipoprotein (HDL) cholesterol has been reported in major depressive disorder, bipolar disorder, first episode of psychosis, and schizophrenia. HDL, its major apolipoprotein component, ApoA1, and the antioxidant enzyme paraoxonase (PON)1 (which is normally bound to ApoA1) all have anti-atherogenic, antioxidant, anti-inflammatory, and immunomodulatory roles, which are discussed in this paper. The paper details the pathways mediating the anti-inflammatory effects of HDL, ApoA1 and PON1 and describes the mechanisms leading to compromised HDL and PON1 levels and function in an environment of chronic inflammation. The molecular mechanisms by which changes in HDL, ApoA1 and PON1 might contribute to the pathophysiology of the neuroprogressive disorders are explained. Moreover, the anti-inflammatory actions of ApoM-mediated sphingosine 1-phosphate (S1P) signalling are reviewed as well as the deleterious effects of chronic inflammation and oxidative stress on ApoM/S1P signalling. Finally, therapeutic interventions specifically aimed at improving the levels and function of HDL and PON1 while reducing levels of inflammation and oxidative stress are considered. These include the so-called Mediterranean diet, extra virgin olive oil, polyphenols, flavonoids, isoflavones, pomegranate juice, melatonin and the Mediterranean diet combined with the ketogenic diet.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | | | - Chiara C Bortolasci
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.
| | - Andre Carvalho
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Estefania G Moreira
- Post-Graduation Program in Health Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
15
|
Marfia G, Navone S, Guarnaccia L, Campanella R, Mondoni M, Locatelli M, Barassi A, Fontana L, Palumbo F, Garzia E, Ciniglio Appiani G, Chiumello D, Miozzo M, Centanni S, Riboni L. Decreased serum level of sphingosine-1-phosphate: a novel predictor of clinical severity in COVID-19. EMBO Mol Med 2021; 13:e13424. [PMID: 33190411 PMCID: PMC7744841 DOI: 10.15252/emmm.202013424] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
The severity of coronavirus disease 2019 (COVID-19) is a crucial problem in patient treatment and outcome. The aim of this study is to evaluate circulating level of sphingosine-1-phosphate (S1P) along with severity markers, in COVID-19 patients. One hundred eleven COVID-19 patients and forty-seven healthy subjects were included. The severity of COVID-19 was found significantly associated with anemia, lymphocytopenia, and significant increase of neutrophil-to-lymphocyte ratio, ferritin, fibrinogen, aminotransferases, lactate dehydrogenase (LDH), C-reactive protein (CRP), and D-dimer. Serum S1P level was inversely associated with COVID-19 severity, being significantly correlated with CRP, LDH, ferritin, and D-dimer. The decrease in S1P was strongly associated with the number of erythrocytes, the major source of plasma S1P, and both apolipoprotein M and albumin, the major transporters of blood S1P. Not last, S1P was found to be a relevant predictor of admission to an intensive care unit, and patient's outcome. Circulating S1P emerged as negative biomarker of severity/mortality of COVID-19 patients. Restoring abnormal S1P levels to a normal range may have the potential to be a therapeutic target in patients with COVID-19.
Collapse
Affiliation(s)
- Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Istituto di Medicina Aerospaziale "A. Mosso"Aeronautica MilitareMilanItaly
- Aldo Ravelli” Research CenterMilanItaly
| | - Stefania Navone
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Aldo Ravelli” Research CenterMilanItaly
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Michele Mondoni
- Respiratory UnitASST Santi Paolo e CarloDepartment of Health SciencesUniversità degli Studi di MilanoMilanItaly
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell TherapyNeurosurgery UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Aldo Ravelli” Research CenterMilanItaly
- Department of Medical‐Surgical Physiopathology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Alessandra Barassi
- Laboratory of Clinical BiochemistryASST Santi Paolo e CarloDepartment of Health SciencesUniversità degli Studi di MilanoMilanItaly
| | - Laura Fontana
- Department of Medical‐Surgical Physiopathology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Fabrizio Palumbo
- Istituto di Medicina Aerospaziale "A. Mosso"Aeronautica MilitareMilanItaly
| | - Emanuele Garzia
- Istituto di Medicina Aerospaziale "A. Mosso"Aeronautica MilitareMilanItaly
- Reproductive Medicine UnitASST Santi Paolo e CarloUniversità degli Studi di MilanoMilanItaly
| | | | | | - Monica Miozzo
- Department of Medical‐Surgical Physiopathology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Unit of Research Laboratories CoordinationFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Stefano Centanni
- Respiratory UnitASST Santi Paolo e CarloDepartment of Health SciencesUniversità degli Studi di MilanoMilanItaly
| | - Laura Riboni
- Department of Medical Biotechnology and Translational MedicineLITA‐Segrate, Università degli Studi di MilanoMilanItaly
| |
Collapse
|
16
|
Yao S, Luo G, Liu H, Zhang J, Zhan Y, Xu N, Zhang X, Zheng L. Apolipoprotein M promotes the anti-inflammatory effect of high-density lipoprotein by binding to scavenger receptor BI. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1676. [PMID: 33490188 PMCID: PMC7812182 DOI: 10.21037/atm-20-7008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Inflammation participates pivotally in the pathogenesis of atherosclerosis. Apolipoprotein M (apoM) is a high-density lipoprotein (HDL)-associated plasma protein that affects HDL metabolism and shows various anti-inflammatory functions in atherosclerosis. In this study, we aim to determine whether apoM is expressed in peripheral blood mononuclear cells (PBMCs) and promoted the anti-inflammatory effect of HDL by combing with scavenger receptor BI (SR-BI). Methods The expression of apoM in PBMCs is detected by a confocal fluorescence microscope and flow cytometry. The interactions between apoM and SR-BI are detected with co-immunoprecipitation. The multiplexed Luminex xMAP assay detects the inflammatory factors induced by apoM+ HDL and apoM– HDL in inflammatory cell model. Results ApoM is expressed on CD14+ monocytes, CD3+ T cells, and CD19+ B cells, CD16+ and CD56+ NK cells. CD14+ monocytes have the highest ratio of apoM+ cells. ApoM+ HDL, apoM– HDL, and recombinant apoM protein could be co-precipitated with SR-BI on the surface of human THP-1 monocytic leukemia cells. In vitro, apoM+ HDL induces significantly less expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-1β than apoM– HDL. Conclusions ApoM was expressed on all PBMCs. ApoM interacted with SR-BI on THP-1. ApoM+ HDL has a more significant anti-inflammatory effect than apoM– HDL.
Collapse
Affiliation(s)
- Shuang Yao
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Guanghua Luo
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hong Liu
- Department of Cardiothoracic Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Zhang
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yuxia Zhan
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ning Xu
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lunds University, Lunds, Sweden
| | - Xiaoying Zhang
- Department of Cardiothoracic Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lu Zheng
- Clinical Medical Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
17
|
Márquez AB, Nazir S, van der Vorst EP. High-Density Lipoprotein Modifications: A Pathological Consequence or Cause of Disease Progression? Biomedicines 2020; 8:biomedicines8120549. [PMID: 33260660 PMCID: PMC7759904 DOI: 10.3390/biomedicines8120549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
High-density lipoprotein (HDL) is well-known for its cardioprotective effects, as it possesses anti-inflammatory, anti-oxidative, anti-thrombotic, and cytoprotective properties. Traditionally, studies and therapeutic approaches have focused on raising HDL cholesterol levels. Recently, it became evident that, not HDL cholesterol, but HDL composition and functionality, is probably a more fruitful target. In disorders, such as chronic kidney disease or cardiovascular diseases, it has been observed that HDL is modified and becomes dysfunctional. There are different modification that can occur, such as serum amyloid, an enrichment and oxidation, carbamylation, and glycation of key proteins. Additionally, the composition of HDL can be affected by changes to enzymes such as cholesterol ester transfer protein (CETP), lecithin-cholesterol acyltransferase (LCAT), and phospholipid transfer protein (PLTP) or by modification to other important components. This review will highlight some main modifications to HDL and discuss whether these modifications are purely a consequential result of pathology or are actually involved in the pathology itself and have a causal role. Therefore, HDL composition may present a molecular target for the amelioration of certain diseases, but more information is needed to determine to what extent HDL modifications play a causal role in disease development.
Collapse
Affiliation(s)
- Andrea Bonnin Márquez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Sumra Nazir
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P.C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, 80336 Munich, Germany
- Correspondence: ; Tel.: +49-241-80-36914
| |
Collapse
|
18
|
Chen Z, Hu M. The apoM-S1P axis in hepatic diseases. Clin Chim Acta 2020; 511:235-242. [PMID: 33096030 DOI: 10.1016/j.cca.2020.10.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
Liver dysfunction is always accompanied by lipid metabolism dysfunction. Apolipoprotein M (apoM), a member of the apolipoprotein family, is primarily expressed and secreted from the liver. apoM is the main chaperone of sphingosine-1-phosphate (S1P), a small signalling molecule associated with numerous physiologic and pathophysiologic processes. In addition to transport, apoM also influences the biologic effects of S1P. Most recently, numerous studies have investigated the potential role of the apoM-S1P axis in a variety of hepatic diseases. These include liver fibrosis, viral hepatitis B and C infection, hepatobiliary disease, non-alcoholic and alcoholic steatohepatitis, acute liver injury and hepatocellular carcinoma. In this review, the roles of apoM and S1P in the development of hepatic diseases are summarized, and novel insights into the diagnosis and treatment of hepatic diseases are discussed.
Collapse
Affiliation(s)
- Zhiyang Chen
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, PR China
| | - Min Hu
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, PR China.
| |
Collapse
|
19
|
Li Y, Zhou J, Qiu J, Huang Z, Wang W, Wu P, Feng A. Berberine reduces gut-vascular barrier permeability via modulation of ApoM/S1P pathway in a model of polymicrobial sepsis. Life Sci 2020; 261:118460. [PMID: 32961234 DOI: 10.1016/j.lfs.2020.118460] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/29/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022]
Abstract
AIMS The hyperpermeability of gut-vascular barrier (GVB) plays a role in gut-derived sepsis. The goal of this study was to evaluate if berberine might improve hepatic apolipoprotein M (ApoM) generation and raise plasma ApoM level to protect the compromised GVB. MATERIALS AND METHODS The compromised GVB was induced by sepsis. Hepatic ApoM mRNA and phosphoenolpyruvate carboxykinase (PEPCK) mRNA and plasma ApoM level were assayed by qRT-PCR and ELISA, respectively. The permeability of intestinal capillary in vivo and of rat intestinal microvascular endothelial cells (RIMECs) in vitro was assayed by FITC-dextran. The blood glucose was detected by a glucometer. Plasma insulin, TNF-α and IL-1β were assayed by ELISA. The plasmalemma vesicle-associated protein-1 (PV1), β-catenin and occludin in RIMECs were assayed by Western blot. KEY FINDINGS Sepsis decreased hepatic ApoM mRNA and plasma ApoM level, but raised hepatic PEPCK mRNA and plasma glucose, insulin, TNF-α, and IL-1β levels. The increased vascular endothelial permeability was abrogated by recombinant rat ApoM in vivo or ApoM-bound S1P in vitro. ApoM-bound S1P decreased PV1 but increased occludin and β-catenin expression in LPS-treated RIMECs. Berberine in a dose-dependent manner raised hepatic ApoM mRNA and plasma ApoM level, but decreased septic hyperglycemia, insulin resistance and plasma TNF-α and IL-1β levels. Berberine reduced sepsis-induced PEPCK and TLR4 mRNA overexpression in the liver. SIGNIFICANCE This study demonstrated berberine inhibited TLR4-mediated hyperglycemia, insulin resistance and proinflammatory molecule production, thereby increasing ApoM gene expression and plasma ApoM. Berberine protected the damaged GVB via modulation of ApoM/S1P pathway.
Collapse
Affiliation(s)
- Yanning Li
- Department of Gastrointestinal Surgery, Maoming People's Hospital, Maoming Clinical Medical College, Guangdong Medical University, Guangdong Province, China
| | - Jun Zhou
- Department of Gastrointestinal Surgery, Maoming People's Hospital, Maoming Clinical Medical College, Guangdong Medical University, Guangdong Province, China
| | - Jiasheng Qiu
- Department of Gastrointestinal Surgery, Maoming People's Hospital, Maoming Clinical Medical College, Guangdong Medical University, Guangdong Province, China
| | - Zudong Huang
- Department of Gastrointestinal Surgery, Maoming People's Hospital, Maoming Clinical Medical College, Guangdong Medical University, Guangdong Province, China
| | - Weiwei Wang
- Department of Gastrointestinal Surgery, Maoming People's Hospital, Maoming Clinical Medical College, Guangdong Medical University, Guangdong Province, China
| | - Ping Wu
- Department of Gastroenterology, Maoming People's Hospital, Maoming Clinical Medical School, Guangdong Medical University, Guangdong Province, China
| | - Aiwen Feng
- Department of Gastrointestinal Surgery, Maoming People's Hospital, Maoming Clinical Medical College, Guangdong Medical University, Guangdong Province, China.
| |
Collapse
|
20
|
Chen Y, Lu Y, Pei C, Liang J, Ding P, Chen S, Hou SZ. Monotropein alleviates secondary liver injury in chronic colitis by regulating TLR4/NF-κB signaling and NLRP3 inflammasome. Eur J Pharmacol 2020; 883:173358. [PMID: 32710952 DOI: 10.1016/j.ejphar.2020.173358] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 01/25/2023]
Abstract
Recently, it has reported that many inflammatory bowel disease (IBD) patients were contracted secondary liver injury. Monotropein (MON), an iridoid glycoside, is demonstrated to possess protective effects on acute colitis mice due to its anti-inflammatory activities. However, it was remained unknown whether MON could inhibit secondary liver injury caused by IBD. The aim of the present study was to investigate the protective roles and mechanisms of MON on secondary liver injury in chronic colitis mice model. In this study, 2% Dextran sodium sulfate (DSS) was used to induce mice model of chronic colitis. The results showed that MON attenuated DSS-induced hepatic pathological damage, liver parameters, infiltration of macrophages and cytokines levels. Furthermore, we found that MON attenuated liver injury through suppressing the activation of the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway and down-regulating the activity of NLRP3 (NOD-, LRR- and pyrin domain-containing 3) inflammasome. All the data indicated that MON may be an effective therapeutic reagent to attenuate secondary liver injury induced by chronic colitis.
Collapse
Affiliation(s)
- Yonger Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yingyu Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Chaoying Pei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jian Liang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Ping Ding
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Shuxian Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, Guangdong, PR China.
| | - Shao-Zhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China; Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| |
Collapse
|
21
|
Cai H, Yao W, Huang J, Xiao J, Chen W, Hu L, Mai R, Liang M, Chen D, Jiang N, Zhou L, Peng T. Apolipoprotein M, identified as a novel hepatitis C virus (HCV) particle associated protein, contributes to HCV assembly and interacts with E2 protein. Antiviral Res 2020; 177:104756. [PMID: 32119870 DOI: 10.1016/j.antiviral.2020.104756] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/18/2020] [Accepted: 02/25/2020] [Indexed: 02/08/2023]
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver diseases such as steatosis, cirrhosis, and hepatocellular carcinoma. HCV particles have been found to associate with apolipoproteins, and apolipoproteins not only participate in the HCV life cycle, but also help HCV escape recognition by the host immune system, which pose challenges for the development of both HCV treatments and vaccines. However, no study has reported on the comprehensive identification of apolipoprotein associations with HCV particles. In the present study, we performed proteome analysis by affinity purification coupled with mass spectrometry (AP-MS) to comprehensively identify the apolipoprotein associations with HCV particles, and ApoM was first identified by AP-MS besides the previously reported ApoE, ApoB, ApoA-I and ApoC-I. Additionally, three assays further confirmed that ApoM was a novel virus particle associated protein. We also showed that ApoM was required for HCV production, especially for the assembly/release step of HCV life cycle. Furthermore, ApoM interacted with the HCV E2 protein. Finally, HCV infection reduced ApoM expression both in vitro and in vivo. Collectively, our study demonstrates that ApoM, identified as a novel HCV particle associated protein, contributes to HCV assembly/release and interacts with HCV E2 protein. It provides new insights on how HCV and the host apolipoproteins are reciprocally influenced and lays a basis for research in developing innovative antiviral strategies.
Collapse
Affiliation(s)
- Hua Cai
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Wenxia Yao
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Jingxian Huang
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jing Xiao
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Wenli Chen
- Department of Infectious Diseases, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Longbo Hu
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Runming Mai
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Mengdi Liang
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Di Chen
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Nan Jiang
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Zhou
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Peng
- Guangzhou Hoffmann Institute of Immunology, College of Basic Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
22
|
Tian Y, Wen H, Qi X, Mao X, Shi Z, Li J, He F, Yang W, Zhang X, Li Y. Analysis of apolipoprotein multigene family in spotted sea bass (Lateolabrax maculatus) and their expression profiles in response to Vibrio harveyi infection. FISH & SHELLFISH IMMUNOLOGY 2019; 92:111-118. [PMID: 31176005 DOI: 10.1016/j.fsi.2019.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 06/09/2023]
Abstract
Apolipoproteins (Apos), which are the protein components of plasma lipoproteins, play important roles in lipid transport in vertebrates. It has been demonstrated that in teleosts, several Apos display antimicrobial activity and play crucial roles in innate immunity. Despite their importance, apo genes have not been systematically characterized in many aquaculture fish species. In our study, a complete set of 23 apo genes was identified and annotated from spotted sea bass (Lateolabrax maculatus). Phylogenetic and homology analyses provided evidence for their annotation and evolutionary relationships. To investigate their potential roles in the immune response, the expression patterns of 23 apo genes were determined in the liver and intestine by qRT-PCR after Vibrio harveyi infection. After infection, a total of 20 differentially expressed apo genes were observed, and their expression profiles varied among the genes and tissues. 5 apo genes (apoA1, apoA4a.1, apoC2, apoF and apoO) were dramatically induced or suppressed (log2 fold change >4, P < 0.05), suggesting their involvement in the immune response of spotted sea bass. Our study provides a valuable foundation for future studies aimed at uncovering the specific roles of each apo gene during bacterial infection in spotted sea bass and other teleost species.
Collapse
Affiliation(s)
- Yuan Tian
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Xin Qi
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Xuebin Mao
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Zhijie Shi
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Jifang Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Feng He
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Wenzhao Yang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Xiaoyan Zhang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China.
| | - Yun Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China.
| |
Collapse
|
23
|
Tydén H, Lood C, Jönsen A, Gullstrand B, Kahn R, Linge P, Kumaraswamy SB, Dahlbäck B, Bengtsson AA. Low plasma concentrations of apolipoprotein M are associated with disease activity and endothelial dysfunction in systemic lupus erythematosus. Arthritis Res Ther 2019; 21:110. [PMID: 31046824 PMCID: PMC6498515 DOI: 10.1186/s13075-019-1890-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/02/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Apolipoprotein M (apoM) is a 25-kDa apolipoprotein present in 5% of high-density lipoprotein (HDL) particles. It is suggested to be anti-atherogenic and to play a key role in sustaining endothelial barrier integrity. SLE patients have increased cardiovascular disease risk, and we aimed to investigate if apoM levels reflect endothelial function in SLE. Since apoM plasma levels decrease during inflammatory conditions, our aim was also to determine the impact of SLE disease activity on apoM plasma levels. METHODS Plasma concentrations of apoM were measured by ELISA in two patient groups with systemic lupus erythematosus (SLE) and in 79 healthy control individuals. In patient group I (n = 84), evaluation time points were selected with the objective to include a wide range of clinical and laboratory variables reflecting disease activity which was measured as SLEDAI. In patient group II consisting of 140 consecutive patients, endothelial function was measured by a finger plethysmograph. A low Reactive Hyperemia Index (RHI) value indicates endothelial dysfunction. RESULTS SLE patients had decreased levels of apoM compared to healthy controls (p < 0.01), with apoM levels correlating inversely with SLEDAI (r = - 0.31, p < 0.01) as well as with levels of CRP (r = - 0.26, p = 0.02) and positively with levels of C3 (r = 0.29, p < 0.01). ApoM levels were particularly low in patients with active disease from the kidney and skin and in patients with leukopenia or positive anti-dsDNA antibody test (p < 0.05). ApoM levels correlated with RHI values in young SLE patients (r = 0.32, p = 0.01), consistent with the important role of apoM in regulating endothelial integrity. CONCLUSIONS ApoM levels may be regulated by SLE-related inflammatory processes and could be a marker of disease activity and endothelial dysfunction, in particular in young SLE patients. Further studies are needed to investigate the predictive value of apoM in the development of a cardiovascular disease.
Collapse
Affiliation(s)
- Helena Tydén
- Department of Rheumatology, Clinical Sciences, Lund University, SE-22185, Lund, Sweden.
| | - Christian Lood
- Department of Rheumatology, Clinical Sciences, Lund University, SE-22185, Lund, Sweden
| | - Andreas Jönsen
- Department of Rheumatology, Clinical Sciences, Lund University, SE-22185, Lund, Sweden
| | - Birgitta Gullstrand
- Department of Rheumatology, Clinical Sciences, Lund University, SE-22185, Lund, Sweden
| | - Robin Kahn
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Petrus Linge
- Department of Rheumatology, Clinical Sciences, Lund University, SE-22185, Lund, Sweden
| | - Sunil B Kumaraswamy
- Department of Translational Medicine, Lund University, 214 28, Malmö, Sweden
| | - Björn Dahlbäck
- Department of Translational Medicine, Lund University, 214 28, Malmö, Sweden
| | - Anders A Bengtsson
- Department of Rheumatology, Clinical Sciences, Lund University, SE-22185, Lund, Sweden
| |
Collapse
|
24
|
Alirezaei F, Ghanbari-Niaki A, Joshaghani H, Naghizadeh Ghomi M. Effect of Aerobic Interval Training on Plasma Apolipoprotein M Levels in Young Men. MEDICAL LABORATORY JOURNAL 2019. [DOI: 10.29252/mlj.13.3.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
25
|
Zheng Z, Zeng Y, Zhu X, Tan Y, Li Y, Li Q, Yi G. ApoM-S1P Modulates Ox-LDL-Induced Inflammation Through the PI3K/Akt Signaling Pathway in HUVECs. Inflammation 2018; 42:606-617. [DOI: 10.1007/s10753-018-0918-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
26
|
Zhu B, Luo GH, Feng YH, Yu MM, Zhang J, Wei J, Yang C, Xu N, Zhang XY. Apolipoprotein M Protects Against Lipopolysaccharide-Induced Acute Lung Injury via Sphingosine-1-Phosphate Signaling. Inflammation 2018; 41:643-653. [PMID: 29260347 DOI: 10.1007/s10753-017-0719-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
It had been demonstrated that apolipoprotein M (apoM) is an important carrier of sphingosine-1-phosphate (S1P) in blood, and the S1P has critical roles in the pathogenesis of sepsis-induced acute lung injury (ALI). In the present study, we investigated whether apoM has beneficial effects in a mouse model after lipopolysaccharide (LPS)-induced ALI. Forty-eight mice were divided into two groups: male C57BL/6 wild-type (apoM+/+) group (n = 24) and apoM gene-deficient (apoM-/-) group (n = 24) and then randomly subdivided into four subgroups (n = 6 each) according to different intraperitoneal (i.p.) injection: control group, W146 group, LPS group, and LPS + W146 group. Serum levels of interleukin-1 beta (IL-1β) and mRNA levels of IL-1β, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), lung histology, wet/dry weight ratio, and immunohistochemistry were measured at 3 h after the baseline and compared in each group. Our results clearly demonstrated that IL-1β mRNA levels and other inflammatory biomarkers were significantly increased in the lungs of LPS-induced ALI apoM-/- mice compared to those of the apoM+/+ mice. Moreover, when apoM+/+ mice were treated with W146, a S1P receptor (S1PR1) antagonist, these inflammatory biomarkers could be significantly upregulated by LPS-induced ALI. Therefore, it suggests that apoM-S1P-S1PR1 signaling might underlie the pathogenesis of ALI and apoM could have physiological benefits to alleviate LPS-induced ALI.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213003, China
| | - Guang-Hua Luo
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213003, China
| | - Yue-Hua Feng
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213003, China
| | - Miao-Mei Yu
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213003, China
| | - Jun Zhang
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213003, China
| | - Jiang Wei
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213003, China
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Ning Xu
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, 221 85, Lund, Sweden
| | - Xiao-Ying Zhang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, Jiangsu Province, 213003, China.
| |
Collapse
|
27
|
Haghikia A, Landmesser U. High-Density Lipoproteins: Effects on Vascular Function and Role in the Immune Response. Cardiol Clin 2018; 36:317-327. [PMID: 29609761 DOI: 10.1016/j.ccl.2017.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The focus in studies of high-density lipoproteins was on their capacity to remove excess cholesterol and deliver it to the liver. Other functions and vascular effects have been described. Clinical trials and translational/genetic studies have led to a refined understanding of the role of high-density lipoprotein; it is likely not a causal cardiovascular risk factor. In healthy subjects, it limits lipid oxidation, protects endothelial cell functions/integrity, and exerts antiinflammatory/antiapoptotic effects. In patients with coronary disease or diabetes, it undergoes modifications/remodeling, resulting in dysfunctional high-density lipoprotein. We summarize recent findings about the regulation of its function and discuss the clinical implications.
Collapse
Affiliation(s)
- Arash Haghikia
- Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin 12203, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Postfach 65 21 33, Berlin 13316, Germany.
| | - Ulf Landmesser
- Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin 12203, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Postfach 65 21 33, Berlin 13316, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, Berlin 10178, Germany
| |
Collapse
|
28
|
Li T, Yang L, Zhao S, Zhang S. Correlation Between Apolipoprotein M and Inflammatory Factors in Obese Patients. Med Sci Monit 2018; 24:5698-5703. [PMID: 30110274 PMCID: PMC6106616 DOI: 10.12659/msm.907744] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background The aim of this study was to observe apolipoprotein M (ApoM) level in obese patients and to explore its correlation with inflammatory factors. Material/Methods A total number of 96 participants were recruited and divided into 2 groups: the control group (or healthy group) whose participants had normal body weight (n=58), and the obese group with all its participants diagnosed with obesity (n=38). Data on blood pressure, body weight, height, body mass index, diastolic function of brachial artery endothelium, fasting venous blood glucose, blood lipids, plasmatic ApoM, interleukin-6 (IL-6), C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), fasting insulin, and adiponectin levels were collected for both groups. Results In the obese group, the levels of plasmatic ApoM, high-density lipoprotein cholesterol (HDL-C), and plasmatic adiponectin were significantly (p<0.05) decreased compared to the control group, and the levels of IL-6, TNF-α, CRP, and fasting insulin were significantly increased (p<0.05) compared to the control group. For the obese group, plasmatic ApoM level was positively correlated with HDL-C level and negatively correlated with levels of IL-6, TNF-α, CRP, insulin, and insulin resistance index. However, no significant correlations were revealed between plasmatic ApoM and the diastolic function of brachial artery endothelium, adiponectin level, blood pressure, and blood glucose level. Conclusions Obese patients showed significantly lower plasmatic ApoM levels than people with normal body weight, and ApoM level showed a strong correlation with CRP, TNF-α, and IL-6 levels, which indicated that ApoM might be regulated by these inflammatory factors.
Collapse
Affiliation(s)
- Tie Li
- Department of Cardiology, Changsha Central Hospital, Changsha, Hunan, China (mainland)
| | - Liu Yang
- Department of Geriatric, International Medical Services, Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Shuiping Zhao
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Saidan Zhang
- Department of Cardiology, Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
29
|
Rohrbach T, Maceyka M, Spiegel S. Sphingosine kinase and sphingosine-1-phosphate in liver pathobiology. Crit Rev Biochem Mol Biol 2017; 52:543-553. [PMID: 28618839 DOI: 10.1080/10409238.2017.1337706] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over 20 years ago, sphingosine-1-phosphate (S1P) was discovered to be a bioactive signaling molecule. Subsequent studies later identified two related kinases, sphingosine kinase 1 and 2, which are responsible for the phosphorylation of sphingosine to S1P. Many stimuli increase sphingosine kinase activity and S1P production and secretion. Outside the cell, S1P can bind to and activate five S1P-specific G protein-coupled receptors (S1PR1-5) to regulate many important cellular and physiological processes in an autocrine or paracrine manner. S1P is found in high concentrations in the blood where it functions to control vascular integrity and trafficking of lymphocytes. Obesity increases blood S1P levels in humans and mice. With the world wide increase in obesity linked to consumption of high-fat, high-sugar diets, S1P is emerging as an accomplice in liver pathobiology, including acute liver failure, metabolic syndrome, control of blood lipid and glucose homeostasis, nonalcoholic fatty liver disease, and liver fibrosis. Here, we review recent research on the importance of sphingosine kinases, S1P, and S1PRs in liver pathobiology, with a focus on exciting insights for new therapeutic modalities that target S1P signaling axes for a variety of liver diseases.
Collapse
Affiliation(s)
- Timothy Rohrbach
- a Department of Biochemistry and Molecular Biology and the Massey Cancer Center , VCU School of Medicine , Richmond , VA , USA
| | - Michael Maceyka
- a Department of Biochemistry and Molecular Biology and the Massey Cancer Center , VCU School of Medicine , Richmond , VA , USA
| | - Sarah Spiegel
- a Department of Biochemistry and Molecular Biology and the Massey Cancer Center , VCU School of Medicine , Richmond , VA , USA
| |
Collapse
|
30
|
Milovanović I, Busarčević M, Trbovich A, Ivović V, Uzelac A, Djurković-Djaković O. Evidence for host genetic regulation of altered lipid metabolism in experimental toxoplasmosis supported with gene data mining results. PLoS One 2017; 12:e0176700. [PMID: 28459857 PMCID: PMC5411058 DOI: 10.1371/journal.pone.0176700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/14/2017] [Indexed: 12/23/2022] Open
Abstract
Toxoplasma gondii is one of the most successful parasites on Earth, infecting a wide array of mammals including one third of the global human population. The obligate intracellular protozoon is not capable of synthesizing cholesterol (Chl), and thus depends on uptake of host Chl for its own development. To explore the genetic regulation of previously observed lipid metabolism alterations during acute murine T. gondii infection, we here assessed total Chl and its fractions in serum and selected tissues at the pathophysiological and molecular level, and integrated the observed gene expression of selected molecules relevant for Chl metabolism, including its biosynthetic and export KEGG pathways, with the results of published transcriptomes obtained in similar murine models of T. gondii infection. The serum lipid status as well as the transcript levels of relevant genes in the brain and the liver were assessed in experimental models of acute and chronic toxoplasmosis in wild-type mice. The results showed that acute infection was associated with a decrease in Chl content in both the liver and periphery (brain, peripheral lymphocytes), and a decrease in Chl reverse transport. In contrast, in chronic infection, a return to normal levels of Chl metabolism has been noted. These changes corresponded to the brain and liver gene expression results as well as to data obtained via mining. We propose that the observed changes in Chl metabolism are part of the host defense response. Further insight into the lipid metabolism in T. gondii infection may provide novel targets for therapeutic agents.
Collapse
Affiliation(s)
- Ivan Milovanović
- Institute for Pathologic Physiology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Miloš Busarčević
- National Reference Laboratory for Toxoplasmosis, Center of Excellence for Food- and Vector-borne Zoonoses, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Alexander Trbovich
- Institute for Pathologic Physiology, School of Medicine, University of Belgrade, Belgrade, Serbia
- National Reference Laboratory for Toxoplasmosis, Center of Excellence for Food- and Vector-borne Zoonoses, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Vladimir Ivović
- National Reference Laboratory for Toxoplasmosis, Center of Excellence for Food- and Vector-borne Zoonoses, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Uzelac
- National Reference Laboratory for Toxoplasmosis, Center of Excellence for Food- and Vector-borne Zoonoses, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Olgica Djurković-Djaković
- National Reference Laboratory for Toxoplasmosis, Center of Excellence for Food- and Vector-borne Zoonoses, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
- * E-mail:
| |
Collapse
|
31
|
Zhang Y, Huang LZ, Yang QL, Liu Y, Zhou X. Correlation analysis between ApoM gene-promoter polymorphisms and coronary heart disease. Cardiovasc J Afr 2017; 27:228-237. [PMID: 27841911 PMCID: PMC5340899 DOI: 10.5830/cvja-2016-001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 01/08/2016] [Indexed: 11/17/2022] Open
Abstract
Objectives: Apolipoprotein M (ApoM), a 25-kDa plasma protein belonging to the lipocalin protein family, is predominantly associated with high-density lipoprotein cholesterol (HDL-C). Studies have suggested ApoM to be important for the formation of pre-β-HDL and to increase cholesterol efflux from macrophage foam cells. The aim of this study was to explore the association of single-nucleotide polymorphisms(SNPs) in the ApoM promoter with coronary atherosclerotic disease (CAD), and the contribution of latent factors. Methods: ApoM was measured in samples from two separate case–control studies, of whom 88 patients developed CAD and 88 were controls. Whole-blood samples from subjects were genotyped by PCR-restriction fragment length polymorphism (PCR-RFLP). Luciferase activities were measured for HepG2 cells with two SNPs, rs805296 (T-778C) and rs940494 (T-855C), and after interfering with or overexpressing the predicted transcription factors. The ability of the SNPs to combine with nucleoproteins was analysed by electophoretic mobility shift assay (EMSA). Results: Mean plasma ApoM concentrations in the CAD and non-CAD groups were 9.58 ± 4.30 and 12.22 ± 6.59 μg/ ml, respectively. Correlation studies of ApoM concentrations with several analytes showed a marked positive correlation with HDL-C, fasting plasma glucose and triglyceride levels. The CC genotype showed lower luciferase activities compared to the TC and TT genotypes. The ApoM-855 mutant-typecould bind to the AP-2α. Interference and overexpression of AP-2 increased and decreased luciferase activities of the wild and mutant types to different degrees. Conclusion:: ApoM may be a biomarker of CAD. ApoM- 855 T→C substitution provides binding sites for AP-2α and reduces ApoM transcription activity.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biochemistry and Molecular Biology, Wan Nan Medical College, Anhui 241000, China.
| | - Li-Zhu Huang
- Clinical Testing and Diagnosis, Experimental Centre of Bengbu Medical College, Anhui 233000, China
| | - Qing-Ling Yang
- Department of Biochemistry and Molecular Biology, Beng Bu Medical College, Anhui 233000, China
| | - Yan Liu
- Clinical Testing and Diagnosis, Experimental Centre of Bengbu Medical College, Anhui 233000, China
| | - Xin Zhou
- Centre for Gene Diagnosis, Zhongnan Hospital, Wuhan University, Wuhan, Peoples' Republic of China
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Studies have shown that chronic inflammatory disorders, such as rheumatoid arthritis, systemic lupus erythematosus, and psoriasis are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism by which inflammation increases cardiovascular disease is likely multifactorial but changes in HDL structure and function that occur during inflammation could play a role. RECENT FINDINGS HDL levels decrease with inflammation and there are marked changes in HDL-associated proteins. Serum amyloid A markedly increases whereas apolipoprotein A-I, lecithin:cholesterol acyltransferase, cholesterol ester transfer protein, paraoxonase 1, and apolipoprotein M decrease. The exact mechanism by which inflammation decreases HDL levels is not defined but decreases in apolipoprotein A-I production, increases in serum amyloid A, increases in endothelial lipase and secretory phospholipase A2 activity, and decreases in lecithin:cholesterol acyltransferase activity could all contribute. The changes in HDL induced by inflammation reduce the ability of HDL to participate in reverse cholesterol transport and protect LDL from oxidation. SUMMARY During inflammation multiple changes in HDL structure occur leading to alterations in HDL function. In the short term, these changes may be beneficial resulting in an increase in cholesterol in peripheral cells to improve host defense and repair but over the long term these changes may increase the risk of atherosclerosis.
Collapse
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section, Department of Veterans Affairs Medical Center, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
33
|
Tayal D, Goswami B, Tyagi S, Chaudhary M, Mallika V. Interaction between dyslipidaemia, oxidative stress and inflammatory response in patients with angiographically proven coronary artery disease. Cardiovasc J Afr 2016; 23:23-7. [PMID: 22331247 PMCID: PMC3721930 DOI: 10.5830/cvja-2010-092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 09/07/2010] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Coronary artery disease (CAD) is emerging as the biggest killer of the 21st century. A number of theories have been postulated to explain the aetiology of atherosclerosis. The present study attempts to elucidate the interaction, if any, between inflammation, oxidative stress and dyslipidaemia in CAD. METHODS A total of 753 patients undergoing angiography were evaluated and 476 were included in the study. The parameters studied included complete lipid profile, and apolipoprotein B, ferritin and nitric oxide (NO) levels. Statistical analysis was carried out to determine the interrelationship between these parameters and the best predictor of CAD risk. Cut-off points were determined from the receiver operating characteristics curves, and the specificity, sensitivity, positive predictive value, negative predictive value, odds ratio and confidence intervals were calculated. RESULTS The levels of the parameters studied increased with the stenotic state and a positive correlation was observed between ferritin, NO and apolipoprotein B. NO emerged as the most reliable predictor of CAD, with an area under the curve of 0.992 and sensitivity and specificity of 97 and 98%, respectively. CONCLUSION Environmental and genetic risk factors for CAD interact in a highly complex manner to initiate the atherosclerotic process. These risk factors should be considered mutually inclusive, not exclusive when devising pharmacological interventions, as multi-factorial risk management is the cornerstone of CAD management.
Collapse
Affiliation(s)
- D Tayal
- Department of Biochemistry, GB Pant Hospital, New Delhi, India
| | | | | | | | | |
Collapse
|
34
|
High apolipoprotein M serum levels correlate with chronic obstructive pulmonary disease. Lipids Health Dis 2016; 15:59. [PMID: 27001252 PMCID: PMC4802916 DOI: 10.1186/s12944-016-0228-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/15/2016] [Indexed: 11/16/2022] Open
Abstract
Background Recently, variations in a component of high-density lipoprotein (HDL), namely apolipoprotein M (apoM), were found to be associated with chronic obstructive pulmonary disease (COPD). The aim of this study was to evaluate the association between apoM and COPD severity. Factors associated with apoM, COPD, or coronary artery disease (CAD) were also assessed. Methods A total of 110 COPD patients and 110 age- and sex-matched non-COPD controls were included. Among them, thirty COPD patients and seven non-COPD controls had CAD. ApoM and pentraxin-3 levels were measured by ELISA. Additionally, the levels of high-sensitivity C-reactive protein (hs-CRP), cholesterol, and triglyceride were assessed using an automatic biochemical analyzer. Results Serum apoM levels increased gradually with COPD severity, with the most prominent apoM elevation observed in very severe COPD cases. In addition, ApoM was correlated with percent-predicted forced expiratory volume in one second (% predicted FEV1) (r = −0.38, P < 0.001), low-density lipoprotein cholesterol (LDL-C) (r = 0.23, P < 0.017), and hs-CRP (r = 0.24, P = 0.01) in COPD patients. Furthermore, apoM was shown to be a risk factor for COPD onset (OR = 1.095, 95 % CI = 1.034–1.160, P = 0.002), but not associated with CAD in COPD patients. Conclusions Serum apoM was elevated in COPD patients and increased gradually with COPD severity. However, there was no association between apoM and CAD development in COPD patients. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0228-1) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Frej C, Linder A, Happonen KE, Taylor FB, Lupu F, Dahlbäck B. Sphingosine 1-phosphate and its carrier apolipoprotein M in human sepsis and in Escherichia coli sepsis in baboons. J Cell Mol Med 2016; 20:1170-81. [PMID: 26990127 PMCID: PMC4882985 DOI: 10.1111/jcmm.12831] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/07/2016] [Indexed: 01/01/2023] Open
Abstract
Sphingosine 1‐phosphate (S1P) is an important regulator of vascular integrity and immune cell migration, carried in plasma by high‐density lipoprotein (HDL)‐associated apolipoprotein M (apoM) and by albumin. In sepsis, the protein and lipid composition of HDL changes dramatically. The aim of this study was to evaluate changes in S1P and its carrier protein apoM during sepsis. For this purpose, plasma samples from both human sepsis patients and from an experimental Escherichia coli sepsis model in baboons were used. In the human sepsis cohort, previously studied for apoM, plasma demonstrated disease‐severity correlated decreased S1P levels, the profile mimicking that of plasma apoM. In the baboons, a similar disease‐severity dependent decrease in plasma levels of S1P and apoM was observed. In the lethal E. coli baboon sepsis, S1P decreased already within 6–8 hrs, whereas the apoM decrease was seen later at 12–24 hrs. Gel filtration chromatography of plasma from severe human or baboon sepsis on Superose 6 demonstrated an almost complete loss of S1P and apoM in the HDL fractions. S1P plasma concentrations correlated with the platelet count but not with erythrocytes or white blood cells. The liver mRNA levels of apoM and apoA1 decreased strongly upon sepsis induction and after 12 hr both were almost completely lost. In conclusion, during septic challenge, the plasma levels of S1P drop to very low levels. Moreover, the liver synthesis of apoM decreases severely and the plasma levels of apoM are reduced. Possibly, the decrease in S1P contributes to the decreased endothelial barrier function observed in sepsis.
Collapse
Affiliation(s)
- Cecilia Frej
- Department of Translational Medicine, Division of Clinical Chemistry, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Adam Linder
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, Skåne University Hospital, Lund, Sweden
| | - Kaisa E Happonen
- Department of Translational Medicine, Division of Clinical Chemistry, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Fletcher B Taylor
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Björn Dahlbäck
- Department of Translational Medicine, Division of Clinical Chemistry, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
36
|
Christensen PM, Liu CH, Swendeman SL, Obinata H, Qvortrup K, Nielsen LB, Hla T, Di Lorenzo A, Christoffersen C. Impaired endothelial barrier function in apolipoprotein M-deficient mice is dependent on sphingosine-1-phosphate receptor 1. FASEB J 2016; 30:2351-9. [PMID: 26956418 DOI: 10.1096/fj.201500064] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/22/2016] [Indexed: 12/25/2022]
Abstract
Apolipoprotein M (ApoM) transports sphingosine-1-phosphate (S1P) in plasma, and ApoM-deficient mice (Apom(-/-)) have ∼50% reduced plasma S1P levels. There are 5 known S1P receptors, and S1P induces adherens junction formation between endothelial cells through the S1P1 receptor, which in turn suppresses vascular leak. Increased vascular permeability is a hallmark of inflammation. The purpose of this study was to explore the relationships between vascular leakage in ApoM deficiency and S1P1 function in normal physiology and in inflammation. Vascular permeability in the lungs was assessed by accumulation of dextran molecules (70 kDa) and was increased ∼40% in Apom(-/-) mice compared to WT (C57Bl6/j) mice. Reconstitution of plasma ApoM/S1P or treatment with an S1P1 receptor agonist (SEW2871) rapidly reversed the vascular leakage to a level similar to that in WT mice, suggesting that it is caused by decreased plasma levels of S1P and reduced S1P1 stimulation. In a carrageenan-induced model of inflammation, Apom(-/-) mice had increased vascular leakage compared with that in WT mice. Adenoviral overexpression of ApoM in Apom(-/-) mice decreased the vascular leakage compared to adenoviral overexpression of green fluorescent protein. The study suggests that vascular leakage of albumin-sized particles in ApoM deficiency is S1P- and S1P1-dependent and this dependency exacerbates the response to inflammatory stimuli.-Christensen, P. M., Liu, C. H., Swendeman, S. L., Obinata, H., Qvortrup, K., Nielsen, L B., Hla, T., Di Lorenzo, A., Christoffersen, C. Impaired endothelial barrier function in apolipoprotein M-deficient mice is dependent on sphingosine-1-phosphate receptor 1.
Collapse
Affiliation(s)
- Pernille M Christensen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Catherine H Liu
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Steven L Swendeman
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Hideru Obinata
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Klaus Qvortrup
- Department of Biomedical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Lars B Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; and
| | - Timothy Hla
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Annarita Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
37
|
Ma X, Zhao JY, Zhao ZL, Ye J, Li SF, Fang HH, Gu MN, Hu YW, Qin ZS. Propofol Attenuates Lipopolysaccharide-Induced Monocyte Chemoattractant Protein-1 Production Through Enhancing apoM and foxa2 Expression in HepG2 Cells. Inflammation 2016; 38:1329-36. [PMID: 25586482 DOI: 10.1007/s10753-014-0104-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) is a cytokine that mediates the influx of cells to sites of inflammation. Our group recently reported that propofol exerted an anti-inflammatory effect and could inhibit lipopolysaccharide (LPS)-induced production of pro-inflammatory cytokines. However, the effect and possible mechanisms of propofol on MCP-1 expression remain unclear. LPS-stimulated HepG2 cells were treated with 50 μM propofol for 0, 6, 12, and 24 h, respectively. The transcript and protein levels were measured by real-time quantitative PCR and Western blot analyses, respectively. We found that propofol markedly decreased both MCP-1 messenger RNA (mRNA) and protein levels in LPS-stimulated HepG2 cells in a time-dependent manner. Expression of apolipoprotein M (apoM) and forkhead box protein A2 (foxa2) was increased by propofol treatment in HepG2 cells. In addition, the inhibitory effect of propofol on MCP-1 expression was significantly abolished by small interfering RNA against apoM and foxa2 in LPS-stimulated HepG2 cells. Propofol attenuates LPS-induced MCP-1 production through enhancing apoM and foxa2 expression in HepG2 cells.
Collapse
Affiliation(s)
- Xin Ma
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Madsen Svarrer EM, Andersen HØ, Helvind M, Slagman MCJ, Navis G, Dullaart RPF, Dahlbäck B, Nielsen LB. Urinary apolipoprotein M as a biomarker of acute kidney injury in children undergoing heart surgery. Biomark Med 2016; 10:81-93. [DOI: 10.2217/bmm.15.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate whether apoM is excreted in urine of children undergoing heart surgery and the potential of apoM as early biomarker of acute kidney injury (AKI). Materials & methods: Urine was collected in children undergoing heart surgery. ApoM was measured with ELISA. U-apoM was characterized by gel filtration chromatography and western blotting. Results: ApoM was excreted into the urine 0–4 h postoperatively as the full-length apoM in particles smaller than plasma HDL. At 0 h, U-apoM predicted AKI with an area under the receiver-operating characteristics curve of 0.70 (p < 0.018). Sensitivity was 0.71 and specificity was 0.68 at a cutoff level at 1.45 nmol/l. Conclusion: ApoM is excreted in the urine of children after cardiac surgery. Its potential as biomarker of AKI deserves exploration.
Collapse
Affiliation(s)
| | - Henrik Ørbæk Andersen
- Department of Cardiothoracic Surgery, Rigshospitalet, University of Copenhagen, Denmark
| | - Morten Helvind
- Department of Cardiothoracic Surgery, Rigshospitalet, University of Copenhagen, Denmark
| | - Maartje CJ Slagman
- Department of Nephrology, University of Groningen & University Medical Center Groningen, Holland
| | - Gerjan Navis
- Department of Nephrology, University of Groningen & University Medical Center Groningen, Holland
| | - Robin PF Dullaart
- Department of Endocrinology, University of Groningen & University Medical Center Groningen, Holland
| | - Björn Dahlbäck
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, University Hospital, Malmö, Sweden
| | - Lars Bo Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Gao JJ, Hu YW, Wang YC, Sha YH, Ma X, Li SF, Zhao JY, Lu JB, Huang C, Zhao JJ, Zheng L, Wang Q. ApoM Suppresses TNF-α-Induced Expression of ICAM-1 and VCAM-1 Through Inhibiting the Activity of NF-κB. DNA Cell Biol 2015; 34:550-6. [PMID: 26057873 DOI: 10.1089/dna.2015.2892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
To explore the anti-inflammatory effect of apolipoprotein M (apoM) on regulation of tumor necrosis factor-α (TNF-α)-induced expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and further investigate the molecular mechanism of apoM in this process. We found that TNF-α could decrease expression of apoM and inhibitor of NF-κB-α (IκBα) in HepG2 cells. Overexpression of apoM caused a significant decrease of ICAM-1 and VCAM-1 expression, while it caused a significant increase of IκBα expression in HepG2 cells. Furthermore, the treatment with TNF-α could increase ICAM-1 and VCAM-1 expression, decrease IκBα protein expression, and increase nuclear factor-κB (NF-κB) activity, and these effects were markedly enhanced by small interfering RNA (siRNA)-mediated silencing of apoM in HepG2 cells. Our findings demonstrated that apoM suppressed TNF-α-induced expression of ICAM-1 and VCAM-1 through inhibiting the activity of NF-κB.
Collapse
Affiliation(s)
- Ji-Juan Gao
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yan-Wei Hu
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yan-Chao Wang
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yan-Hua Sha
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Xin Ma
- 2 Department of Anesthesiology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Shu-Fen Li
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Jia-Yi Zhao
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Jing-Bo Lu
- 3 Department of Vascular Surgery, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Chuan Huang
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Jing-Jing Zhao
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Lei Zheng
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Qian Wang
- 1 Laboratory Medicine Center, Nanfang Hospital, Southern Medical University , Guangzhou, China
| |
Collapse
|
40
|
Ren K, Tang ZL, Jiang Y, Tan YM, Yi GH. Apolipoprotein M. Clin Chim Acta 2015; 446:21-9. [DOI: 10.1016/j.cca.2015.03.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
|
41
|
Kardassis D, Gafencu A, Zannis VI, Davalos A. Regulation of HDL genes: transcriptional, posttranscriptional, and posttranslational. Handb Exp Pharmacol 2015; 224:113-179. [PMID: 25522987 DOI: 10.1007/978-3-319-09665-0_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
HDL regulation is exerted at multiple levels including regulation at the level of transcription initiation by transcription factors and signal transduction cascades; regulation at the posttranscriptional level by microRNAs and other noncoding RNAs which bind to the coding or noncoding regions of HDL genes regulating mRNA stability and translation; as well as regulation at the posttranslational level by protein modifications, intracellular trafficking, and degradation. The above mechanisms have drastic effects on several HDL-mediated processes including HDL biogenesis, remodeling, cholesterol efflux and uptake, as well as atheroprotective functions on the cells of the arterial wall. The emphasis is on mechanisms that operate in physiologically relevant tissues such as the liver (which accounts for 80% of the total HDL-C levels in the plasma), the macrophages, the adrenals, and the endothelium. Transcription factors that have a significant impact on HDL regulation such as hormone nuclear receptors and hepatocyte nuclear factors are extensively discussed both in terms of gene promoter recognition and regulation but also in terms of their impact on plasma HDL levels as was revealed by knockout studies. Understanding the different modes of regulation of this complex lipoprotein may provide useful insights for the development of novel HDL-raising therapies that could be used to fight against atherosclerosis which is the underlying cause of coronary heart disease.
Collapse
Affiliation(s)
- Dimitris Kardassis
- Department of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology of Hellas, Heraklion, Crete, 71110, Greece,
| | | | | | | |
Collapse
|
42
|
Abstract
During infection significant alterations in lipid metabolism and lipoprotein composition occur. Triglyceride and VLDL cholesterol levels increase, while reduced HDL cholesterol (HDL-C) and LDL cholesterol (LDL-C) levels are observed. More importantly, endotoxemia modulates HDL composition and size: phospholipids are reduced as well as apolipoprotein (apo) A-I, while serum amyloid A (SAA) and secretory phospholipase A2 (sPLA2) dramatically increase, and, although the total HDL particle number does not change, a significant decrease in the number of small- and medium-size particles is observed. Low HDL-C levels inversely correlate with the severity of septic disease and associate with an exaggerated systemic inflammatory response. HDL, as well as other plasma lipoproteins, can bind and neutralize Gram-negative bacterial lipopolysaccharide (LPS) and Gram-positive bacterial lipoteichoic acid (LTA), thus favoring the clearance of these products. HDLs are emerging also as a relevant player during parasitic infections, and a specific component of HDL, namely, apoL-1, confers innate immunity against trypanosome by favoring lysosomal swelling which kills the parasite. During virus infections, proteins associated with the modulation of cholesterol bioavailability in the lipid rafts such as ABCA1 and SR-BI have been shown to favor virus entry into the cells. Pharmacological studies support the benefit of recombinant HDL or apoA-I mimetics during bacterial infection, while apoL-1-nanobody complexes were tested for trypanosome infection. Finally, SR-BI antagonism represents a novel and forefront approach interfering with hepatitis C virus entry which is currently tested in clinical studies. From the coming years, we have to expect new and compelling observations further linking HDL to innate immunity and infections.
Collapse
|
43
|
Abstract
During infections or acute conditions high-density lipoproteins cholesterol (HDL-C) levels decrease very rapidly and HDL particles undergo profound changes in their composition and function. These changes are associated with poor prognosis following endotoxemia or sepsis and data from genetically modified animal models support a protective role for HDL. The same is true for some parasitic infections, where the key player appears to be a specific and minor component of HDL, namely apoL-1. The ability of HDL to influence cholesterol availability in lipid rafts in immune cells results in the modulation of toll-like receptors, MHC-II complex, as well as B- and T-cell receptors, while specific molecules shuttled by HDL such as sphingosine-1-phosphate (S1P) contribute to immune cells trafficking. Animal models with defects associated with HDL metabolism and/or influencing cell cholesterol efflux present features related to immune disorders. All these functions point to HDL as a platform integrating innate and adaptive immunity. The aim of this review is to provide an overview of the connection between HDL and immunity in atherosclerosis and beyond.
Collapse
Affiliation(s)
- Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, via Balzaretti 9, Milan 20133, Italy IRCCS Multimedica, Milan, Italy
| | - Angela Pirillo
- IRCCS Multimedica, Milan, Italy Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, via Balzaretti 9, Milan 20133, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, via Balzaretti 9, Milan 20133, Italy Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy The Blizard Institute, Centre for Diabetes, Barts and The London School of Medicine & Dentistry, Queen Mary University, London, UK
| |
Collapse
|
44
|
Ahmad Y, Sharma NK, Ahmad MF, Sharma M, Garg I, Bhargava K. Proteomic identification of novel differentiation plasma protein markers in hypobaric hypoxia-induced rat model. PLoS One 2014; 9:e98027. [PMID: 24842778 PMCID: PMC4026414 DOI: 10.1371/journal.pone.0098027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/28/2014] [Indexed: 12/24/2022] Open
Abstract
Background Hypobaric hypoxia causes complex changes in the expression of genes, including stress related genes and corresponding proteins that are necessary to maintain homeostasis. Whereas most prior studies focused on single proteins, newer methods allowing the simultaneous study of many proteins could lead to a better understanding of complex and dynamic changes that occur during the hypobaric hypoxia. Methods In this study we investigated the temporal plasma protein alterations of rat induced by hypobaric hypoxia at a simulated altitude of 7620 m (25,000 ft, 282 mm Hg) in a hypobaric chamber. Total plasma proteins collected at different time points (0, 6, 12 and 24 h), separated by two-dimensional electrophoresis (2-DE) and identified using matrix assisted laser desorption ionization time of flight (MALDI-TOF/TOF). Biological processes that were enriched in the plasma proteins during hypobaric hypoxia were identified using Gene Ontology (GO) analysis. According to their properties and obvious alterations during hypobaric hypoxia, changes of plasma concentrations of Ttr, Prdx-2, Gpx -3, Apo A-I, Hp, Apo-E, Fetub and Nme were selected to be validated by Western blot analysis. Results Bioinformatics analysis of 25 differentially expressed proteins showed that 23 had corresponding candidates in the database. The expression patterns of the eight selected proteins observed by Western blot were in agreement with 2-DE results, thus confirming the reliability of the proteomic analysis. Most of the proteins identified are related to cellular defense mechanisms involving anti-inflammatory and antioxidant activity. Their presence reflects the consequence of serial cascades initiated by hypobaric hypoxia. Conclusion/Significance This study provides information about the plasma proteome changes induced in response to hypobaric hypoxia and thus identification of the candidate proteins which can act as novel biomarkers.
Collapse
Affiliation(s)
- Yasmin Ahmad
- Peptide and Proteomics Division, DIPAS, DRDO, Ministry of Defence, Delhi, India
- * E-mail:
| | - Narendra K. Sharma
- Peptide and Proteomics Division, DIPAS, DRDO, Ministry of Defence, Delhi, India
| | | | - Manish Sharma
- Peptide and Proteomics Division, DIPAS, DRDO, Ministry of Defence, Delhi, India
| | - Iti Garg
- Department of Genomics, DIPAS, DRDO, Ministry of Defence, Delhi, India
| | - Kalpana Bhargava
- Peptide and Proteomics Division, DIPAS, DRDO, Ministry of Defence, Delhi, India
| |
Collapse
|
45
|
Urinary apolipoprotein M could be used as a biomarker of acute renal injury: an ischemia-reperfusion injury model of kidney in rat. Transplant Proc 2014; 45:2476-9. [PMID: 23953565 DOI: 10.1016/j.transproceed.2013.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 02/20/2013] [Accepted: 04/23/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND It has been well documented that apolipoprotein M (apoM) is principally expressed in hepatocytes as well as renal tubular epithelial cells. The importance of apoM in the kidney is unknown. In the present study we examined urinary any apoM after short-term ischemia-reperfusion injury (IRI) of kidney in a rat model. METHODS The kidneys of 11 male Sprague-Dawley rats were rendered ischemic for 45 minutes followed by different intervals of reperfusion. Serum and urine apoM concentrations were determined using a dot-blot analysis with specific rabbit anti-human apoM antibodies that cross-react with rat apoM. Serum concentrations of blood urea nitrogen (BUN) and creatinine (Cr) were determined using standard clinical automated analyses. RESULTS BUN was significantly elevated after 45 minutes of ischemia followed by 24 hours of reperfusion; serum Cr concentrations were also significantly increased at 6 and 24 hours of reperfusion. Interestingly, similar to BUN and Cr, serum apoM concentrations were significantly increased after ischemia for 45 minutes alone and after 2 hours of reperfusion. Urinary apoM concentrations were obviously increased after 2 h as well as 6 hours of reperfusion. CONCLUSION apoM showed characteristics of an acute-phase reactive protein; its occurrence in urine may be considered to be a biomarker of acute renal injury.
Collapse
|
46
|
Nojiri T, Kurano M, Tokuhara Y, Ohkubo S, Hara M, Ikeda H, Tsukamoto K, Yatomi Y. Modulation of sphingosine-1-phosphate and apolipoprotein M levels in the plasma, liver and kidneys in streptozotocin-induced diabetic mice. J Diabetes Investig 2014; 5:639-48. [PMID: 25422763 PMCID: PMC4234226 DOI: 10.1111/jdi.12232] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 02/24/2014] [Accepted: 03/16/2014] [Indexed: 12/19/2022] Open
Abstract
AIMS/INTRODUCTION Sphingosine-1-phosphate (S1P), a multifunctional bioactive lipid mediator, is involved in various diseases. Apolipoprotein M (ApoM) carries S1P on high-density lipoprotein and modulates S1P metabolism to increase the total S1P mass in the body. Both S1P and ApoM are involved in diabetes. MATERIALS AND METHODS The present study examined the modulation of S1P and ApoM levels in the plasma, liver and kidneys in streptozotocin-induced diabetes (STZ) mice, and the effects of insulin on the S1P and ApoM levels in the plasma and liver in STZ mice and normal mice. We also examined the effects of insulin and glucose on the ApoM levels in HepG2 cells. RESULTS In STZ mice, both the plasma S1P and ApoM levels were higher than those in control mice. The hepatic S1P and ApoM contents were also elevated. The hepatic S1P and ApoM contents were reduced by insulin treatment, whereas high-dose insulin decreased the plasma S1P and ApoM levels. In mice without streptozotocin treatment, the administration of insulin decreased the plasma S1P and ApoM levels, and the hepatic content of ApoM, whereas the hepatic level of S1P was not altered. Treatment with insulin and incubation under a low glucose level decreased the ApoM levels in HepG2 cells. Regarding the kidney, the renal levels of S1P and ApoM were increased in STZ mice, and insulin treatment partially restored this increment. CONCLUSIONS In STZ mice, the levels of S1P and ApoM in the plasma, liver, and kidneys were increased. Insulin treatment somehow reversed this modulation in STZ mice.
Collapse
Affiliation(s)
- Takahiro Nojiri
- Department of Clinical Laboratory, The University of Tokyo Hospital Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo Tokyo, Japan
| | - Yasunori Tokuhara
- Department of Clinical Laboratory, The University of Tokyo Hospital Tokyo, Japan ; The Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine, Osaka University Osaka, Japan
| | - Shigeo Ohkubo
- Department of Clinical Laboratory, The University of Tokyo Hospital Tokyo, Japan
| | - Masumi Hara
- Department of Medicine IV, Mizonokuchi Hospital, Teikyo University School of Medicine Kawasaki, Japan
| | - Hitoshi Ikeda
- Department of Clinical Laboratory, The University of Tokyo Hospital Tokyo, Japan ; Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo Tokyo, Japan
| | - Kazuhisa Tsukamoto
- Department of Metabolism, Diabetes and Nephrology, Aizu Medical Center, Fukushima Medical University Fukushima, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory, The University of Tokyo Hospital Tokyo, Japan ; Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo Tokyo, Japan
| |
Collapse
|
47
|
Zhao JY, Hu YW, Li SF, Hu YR, Ma X, Wu SG, Wang YC, Gao JJ, Sha YH, Zheng L, Wang Q. Dihydrocapsaicin down-regulates apoM expression through inhibiting Foxa2 expression and enhancing LXRα expression in HepG2 cells. Lipids Health Dis 2014; 13:50. [PMID: 24642298 PMCID: PMC3999941 DOI: 10.1186/1476-511x-13-50] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 03/13/2014] [Indexed: 11/27/2022] Open
Abstract
Background Apolipoprotein M (apoM), as a novel apolipoprotein which is mainly expressed in liver and kidney tissues, is associated with development and progression of atherosclerosis and diabetes. Our group have recently shown that Dihydrocapsaicin(DHC)can significantly decrease atherosclerotic plaque formation in apoE−/− mice. However, the effect and possible mechanism of DHC on apoM expression remain unclear. Methods HepG2 cells were treated with 0 μM, 25 μM, 50 μM and 100 μM DHC for 24 h or were treated with 100 μM DHC for 0, 6, 12, and 24 h, respectively. The mRNA levels and protein levels were measured by real-time quantitative PCR and western blot analysis, respectively. Results We found that DHC markedly decreased expression of apoM at both mRNA and protein level in HepG2 cells in a dose-dependent and time-dependent manner. Expression of Foxa2 was decreased while expression of LXRα was increased by DHC treatment in HepG2 cells. In addittion, overexpression of Foxa2 markedly compensated the inhibition effect induced by DHC on apoM expression. LXRα small interfering RNA significantly abolished the inhibition effect which induced by DHC on apoM expression. The liver of C57BL/6 mice treated with DHC had significantly lower expression of apoM. Furthermore, the liver had lower expression of Foxa2 while had higher expression of LXRα. Conclusions DHC could down-regulate apoM expression through inhibiting Foxa2 expression and enhancing LXRα expression in HepG2 cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | | |
Collapse
|
48
|
Shrestha S, Irvin MR, Grunfeld C, Arnett DK. HIV, inflammation, and calcium in atherosclerosis. Arterioscler Thromb Vasc Biol 2013; 34:244-50. [PMID: 24265418 DOI: 10.1161/atvbaha.113.302191] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is consistently higher among the HIV-positive patients, with or without treatment, than among the HIV-negative population. Risk factors linked to atherosclerotic cardiovascular disease in HIV infection are both traditional and HIV specific although the underlying mechanisms are not fully delineated. Three key sequential biological processes are postulated to accelerate progression of atherosclerosis in the context of HIV: (1) inflammation, (2) transformation of monocytes to macrophages and then foam cells, and (3) apoptosis of foam cells leading to plaque development through Ca(2+)-dependent endoplasmic reticulum stress. These proatherogenic mechanisms are further affected when HIV interacts with the genes involved in various phases within this network.
Collapse
Affiliation(s)
- Sadeep Shrestha
- From the Department of Epidemiology, School of Public Health, University of Alabama at Birmingham (S.S., M.R.I., D.K.A.); Division of Endocrinology and Metabolism, University of California, San Francisco (C.G.); and Veterans Affairs Medical Center, San Francisco, CA (C.G.)
| | | | | | | |
Collapse
|
49
|
Ma X, Hu YW, Zhao ZL, Zheng L, Qiu YR, Huang JL, Wu XJ, Mao XR, Yang J, Zhao JY, Li SF, Gu MN, Wang Q. Anti-inflammatory effects of propofol are mediated by apolipoprotein M in a hepatocyte nuclear factor-1α-dependent manner. Arch Biochem Biophys 2013; 533:1-10. [DOI: 10.1016/j.abb.2013.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 03/01/2013] [Accepted: 03/04/2013] [Indexed: 10/27/2022]
|
50
|
Kumaraswamy SB, Linder A, Åkesson P, Dahlbäck B. Decreased plasma concentrations of apolipoprotein M in sepsis and systemic inflammatory response syndromes. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:R60. [PMID: 22512779 PMCID: PMC3681389 DOI: 10.1186/cc11305] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 03/22/2012] [Accepted: 04/18/2012] [Indexed: 12/14/2022]
Abstract
Introduction Apolipoprotein M (apoM) is present in 5% of high-density lipoprotein (HDL) particles in plasma. It is a carrier of sphingosine-1-phosphate (S1P), which is important for vascular barrier protection. The aim was to determine the plasma concentrations of apoM during sepsis and systemic inflammatory response syndrome (SIRS) and correlate them to levels of apolipoprotein A-I (apoA1), apolipoprotein B (apoB), HDL-, and low-density lipoprotein (LDL)-cholesterol. Methods Plasma samples from patients with (1), severe sepsis with shock (n = 26); (2), severe sepsis without shock (n = 44); (3), sepsis (n = 100); (4), infections without SIRS (n = 43); and (5) SIRS without infection (n = 20) were analyzed. The concentrations of apoM, apoA1, and apoB were measured with enzyme-linked immunosorbent assays (ELISAs). Total, HDL-, and LDL-cholesterol concentrations were measured with a commercial HDL/LDL cholesterol test. Results ApoM concentrations correlated negatively to acute-phase markers. Thus, apoM behaved as a negative acute-phase protein. Decreased values were observed in all patient groups (P < 0.0001), with the most drastic decreases observed in the severely sick patients. ApoM levels correlated strongly to those of apoA1, apoB, HDL, and LDL cholesterol. The HDL and LDL cholesterol levels were low in all patient groups, as compared with controls (P < 0.0001), in particular, HDL cholesterol. ApoA1 and apoB concentrations were low only in the more severely affected patients. Conclusions During sepsis and SIRS, the plasma concentrations of apoM decrease dramatically, the degree of decrease reflecting the severity of the disease. As a carrier for barrier-protective S1P in HDL, the decrease in apoM could contribute to the increased vascular leakage observed in sepsis and SIRS.
Collapse
Affiliation(s)
- Sunil B Kumaraswamy
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Skåne University Hospital, Entrance 46, Malmö, SE-20502, Sweden
| | | | | | | |
Collapse
|