1
|
Liu Q, Zhang D, Liang F, Liu F, Xiao L, An X, Chen X, Liang X. Air pollution and hypertension in rural versus urban children: Lipidomic insights into PM2.5 impacts. ENVIRONMENTAL RESEARCH 2025; 278:121715. [PMID: 40306456 DOI: 10.1016/j.envres.2025.121715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
Fine particulate matter and its impact on blood pressure (BP) in children remain a concern, with the role of lipid metabolism as a potential mediator not fully elucidated. We conducted a cohort study of 2239 urban subjects and 2194 rural subjects at baseline in China from 2014 to 2024 and a nested case-control study with lipidomics analyses. Analysis results showed that higher fine particulate matter smaller than 2.5 μm (PM2.5) exposure associated with high density lipoprotein cholesterol (HDL-C), non-HDL-C, and higher systolic blood pressure (SBP), partially mediated by HDL-C/non-HDL-C changes. Mediation analysis indicated a significant mediating effect of HDL-C on the PM2.5-DBP and PM2.5-MAP (DBP, diastolic blood pressure; MAP, mean arterial pressure) association in urban subjects, while no mediation effect was found in rural subjects. For non-HDL-C, significant mediating effects were observed in both urban and rural subjects. Further analyses revealed distinct urban-rural lipidomic patterns, with specific phosphatidylethanolamine (PEs) mediating PM2.5-related hypertension in rural subjects, while lactosylceramides (LacCer) played this role in urban youth. These patterns extended to other BP indices as well. In the urban area, PG(44:11), LacCer(d45:1), were identified as playing significant mediating roles in the association between PM2.5 exposure and hypertension while for rural subjects, PEs including PE(16:0/16:0) and PE(18:0/18:2) showed significant mediating effects. Our findings underscore the impact of PM2.5 exposure on lipid profiles and BP risk in children, suggesting area-specific mechanisms and the potential for lipidomic-based interventions to mitigate environmental health risks.
Collapse
Affiliation(s)
- Qin Liu
- Department of Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Di Zhang
- Department of Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Fengchao Liang
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Fangchao Liu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leyuan Xiao
- Department of Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Xizhou An
- Department of Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Xin Chen
- Department of Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Xiaohua Liang
- Department of Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| |
Collapse
|
2
|
Kang H, Song J, Cheng Y. HDL regulates the risk of cardiometabolic and inflammatory-related diseases: Focusing on cholesterol efflux capacity. Int Immunopharmacol 2024; 138:112622. [PMID: 38971111 DOI: 10.1016/j.intimp.2024.112622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Dyslipidemia, characterized by higher serum concentrations of low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglyceride (TG), and lower serum concentrations of high-density lipoprotein cholesterol (HDL-C), is confirmed as a hallmark of cardiovascular diseases (CVD), posing serious risks to the future health of humans. Aside from the role of HDL-C concentrations, the capacity of cholesterol efflux to HDL is being identified as an enssential messurement for the dyslipidemic morbidity. Through inducing the progression of reverse cholesterol transport (RCT), the HDL-related cholesterol efflux plays a vital role in atherosclerotic plaque formation. In addition, increasing results demonstrated that the relationships between cholesterol efflux and cardiovascular events might be influenced by multiple factors, such as atherosclerosis, diabetes, and, inflammatory diseases. These risk factors could affect the intracellular composition of HDL, which might subsqently influence the cholesterol efflux process induced by HDL particle. In the present comprehensive article, we summarize the latest findings which described the modulatory roles of HDL in cardiometabolic disorders and inflammatory related diseases, focusing on its capacity in mediating cholesterol efflux. Moreover, the potential mechanisms whereby HDL regulate the risk of cardiometabolic disorders or inflammatory related diseases, at least partly, via cholesterol efflux pathway, are also well-listed.
Collapse
Affiliation(s)
- Huiyuan Kang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jingjin Song
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ye Cheng
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
3
|
Fish-Low CY, Than LTL, Ling KH, Sekawi Z. The Potential of Eight Plasma Proteins as Biomarkers in Redefining Leptospirosis Diagnosis. J Proteome Res 2024; 23:4027-4042. [PMID: 39150348 DOI: 10.1021/acs.jproteome.4c00376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Leptospirosis, a notifiable endemic disease in Malaysia, has higher mortality rates than regional dengue fever. Diverse clinical symptoms and limited diagnostic methods complicate leptospirosis diagnosis. The demand for accurate biomarker-based diagnostics is increasing. This study investigated the plasma proteome of leptospirosis patients with leptospiraemia and seroconversion compared with dengue patients and healthy subjects using isobaric tags for relative and absolute quantitation (iTRAQ)-mass spectrometry (MS). The iTRAQ analysis identified a total of 450 proteins, which were refined to a list of 290 proteins through a series of exclusion criteria. Differential expression in the plasma proteome of leptospirosis patients compared to the control groups identified 11 proteins, which are apolipoprotein A-II (APOA2), C-reactive protein (CRP), fermitin family homolog 3 (FERMT3), leucine-rich alpha-2-glycoprotein 1 (LRG1), lipopolysaccharide-binding protein (LBP), myosin-9 (MYH9), platelet basic protein (PPBP), platelet factor 4 (PF4), profilin-1 (PFN1), serum amyloid A-1 protein (SAA1), and thrombospondin-1 (THBS1). Following a study on a verification cohort, a panel of eight plasma protein biomarkers was identified for potential leptospirosis diagnosis: CRP, LRG1, LBP, MYH9, PPBP, PF4, SAA1, and THBS1. In conclusion, a panel of eight protein biomarkers offers a promising approach for leptospirosis diagnosis, addressing the limitations of the "one disease, one biomarker" concept.
Collapse
Affiliation(s)
- Cheng-Yee Fish-Low
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - Leslie Thian Lung Than
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - King-Hwa Ling
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| |
Collapse
|
4
|
Ji A, Trumbauer AC, Noffsinger VP, Meredith LW, Dong B, Wang Q, Guo L, Li X, De Beer FC, Webb NR, Tannock LR, Starr ME, Waters CM, Shridas P. Deficiency of Acute-Phase Serum Amyloid A Exacerbates Sepsis-Induced Mortality and Lung Injury in Mice. Int J Mol Sci 2023; 24:17501. [PMID: 38139330 PMCID: PMC10744229 DOI: 10.3390/ijms242417501] [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: 11/10/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Serum amyloid A (SAA) is a family of proteins, the plasma levels of which may increase >1000-fold in acute inflammatory states. We investigated the role of SAA in sepsis using mice deficient in all three acute-phase SAA isoforms (SAA-TKO). SAA deficiency significantly increased mortality rates in the three experimental sepsis mouse models: cecal ligation and puncture (CLP), cecal slurry (CS) injection, and lipopolysaccharide (LPS) treatments. SAA-TKO mice had exacerbated lung pathology compared to wild-type (WT) mice after CLP. A bulk RNA sequencing performed on lung tissues excised 24 h after CLP indicated significant enrichment in the expression of genes associated with chemokine production, chemokine and cytokine-mediated signaling, neutrophil chemotaxis, and neutrophil migration in SAA-TKO compared to WT mice. Consistently, myeloperoxidase activity and neutrophil counts were significantly increased in the lungs of septic SAA-TKO mice compared to WT mice. The in vitro treatment of HL-60, neutrophil-like cells, with SAA or SAA bound to a high-density lipoprotein (SAA-HDL), significantly decreased cellular transmigration through laminin-coated membranes compared to untreated cells. Thus, SAA potentially prevents neutrophil transmigration into injured lungs, thus reducing exacerbated tissue injury and mortality. In conclusion, we demonstrate for the first time that endogenous SAA plays a protective role in sepsis, including ameliorating lung injury.
Collapse
Affiliation(s)
- Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Andrea C. Trumbauer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Victoria P. Noffsinger
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Luke W. Meredith
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Brittany Dong
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
| | - Qian Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Ling Guo
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Xiangan Li
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Frederick C. De Beer
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Nancy R. Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Lisa R. Tannock
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Marlene E. Starr
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M. Waters
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
| | - Preetha Shridas
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
5
|
Iyer DR, Venkatraman J, Tanguy E, Vitale N, Mahapatra NR. Chromogranin A and its derived peptides: potential regulators of cholesterol homeostasis. Cell Mol Life Sci 2023; 80:271. [PMID: 37642733 PMCID: PMC11072126 DOI: 10.1007/s00018-023-04908-3] [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: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Chromogranin A (CHGA), a member of the granin family of proteins, has been an attractive therapeutic target and candidate biomarker for several cardiovascular, neurological, and inflammatory disorders. The prominence of CHGA stems from the pleiotropic roles of several bioactive peptides (e.g., catestatin, pancreastatin, vasostatins) generated by its proteolytic cleavage and by their wide anatomical distribution. These peptides are emerging as novel modulators of cardiometabolic diseases that are often linked to high blood cholesterol levels. However, their impact on cholesterol homeostasis is poorly understood. The dynamic nature of cholesterol and its multitudinous roles in almost every aspect of normal body function makes it an integral component of metabolic physiology. A tightly regulated coordination of cholesterol homeostasis is imperative for proper functioning of cellular and metabolic processes. The deregulation of cholesterol levels can result in several pathophysiological states. Although studies till date suggest regulatory roles for CHGA and its derived peptides on cholesterol levels, the mechanisms by which this is achieved still remain unclear. This review aims to aggregate and consolidate the available evidence linking CHGA with cholesterol homeostasis in health and disease. In addition, we also look at common molecular regulatory factors (viz., transcription factors and microRNAs) which could govern the expression of CHGA and genes involved in cholesterol homeostasis under basal and pathological conditions. In order to gain further insights into the pathways mediating cholesterol regulation by CHGA/its derived peptides, a few prospective signaling pathways are explored, which could act as primers for future studies.
Collapse
Affiliation(s)
- Dhanya R Iyer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Janani Venkatraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France.
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
6
|
Adorni MP, Papotti B, Borghi MO, Raschi E, Zimetti F, Bernini F, Meroni PL, Ronda N. Effect of the JAK/STAT Inhibitor Tofacitinib on Macrophage Cholesterol Metabolism. Int J Mol Sci 2023; 24:12571. [PMID: 37628747 PMCID: PMC10454555 DOI: 10.3390/ijms241612571] [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: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The impact of JAK/STAT inhibitors, which are used in various inflammatory diseases, on cardiovascular risk is controversial and has recently raised safety concerns. Our study investigates the direct effects of tofacitinib on macrophage cholesterol metabolism, which is crucial for atherosclerosis plaque development and stability. Cultured human macrophages THP-1 were used to assess the impact of tofacitinib on cell cholesterol efflux and synthesis via radioisotopic methods, and on cholesterol uptake by measuring the cell cholesterol content with a fluorometric assay. The cholesterol acceptors and donors were either standard lipoproteins or sera from patients with juvenile idiopathic arthritis (JIA) and from control subjects. Tofacitinib significantly increased the macrophage cholesterol efflux to all acceptors; it reduced cholesterol uptake from both the normal and hypercholesterolemic sera; and it reduced cholesterol synthesis. The treatment of macrophages with tofacitinib was able to increase the cholesterol efflux and decrease cholesterol uptake when using sera from untreated JIA patients with active disease as cholesterol acceptors and donors, respectively. In conclusion, our in vitro data support the concept that tofacitinib has a favorable impact on macrophage cholesterol metabolism, even in the presence of sera from rheumatologic patients, and suggest that other mechanisms may be responsible for the cardiovascular risk associated with tofacitinib use in selected patient populations.
Collapse
Affiliation(s)
- Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Via Volturno 39/F, 43125 Parma, Italy;
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (B.P.); (F.Z.); (F.B.)
| | - Maria Orietta Borghi
- Experimental Laboratory of Immuno-Rheumatologic Researches, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Via Zucchi 18, 20095 Milan, Italy; (M.O.B.); (E.R.); (P.L.M.)
| | - Elena Raschi
- Experimental Laboratory of Immuno-Rheumatologic Researches, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Via Zucchi 18, 20095 Milan, Italy; (M.O.B.); (E.R.); (P.L.M.)
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (B.P.); (F.Z.); (F.B.)
| | - Franco Bernini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (B.P.); (F.Z.); (F.B.)
| | - Pier Luigi Meroni
- Experimental Laboratory of Immuno-Rheumatologic Researches, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Via Zucchi 18, 20095 Milan, Italy; (M.O.B.); (E.R.); (P.L.M.)
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (B.P.); (F.Z.); (F.B.)
| |
Collapse
|
7
|
Perrone F, Favari E, Maglietta G, Verzè M, Pluchino M, Minari R, Sabato R, Mazzaschi G, Ronca A, Rossi A, Cortellini A, Pecci F, Cantini L, Bersanelli M, Quaini F, Tiseo M, Buti S. The role of blood cholesterol quality in patients with advanced cancer receiving immune checkpoint inhibitors. Cancer Immunol Immunother 2023; 72:2127-2135. [PMID: 36828963 PMCID: PMC10992484 DOI: 10.1007/s00262-023-03398-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 02/04/2023] [Indexed: 02/26/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) became the standard of care for several solid tumors. A limited fraction of patients (pts) achieves a long-term benefit. Plasmatic and intracellular cholesterol levels have emerged as promising biomarkers. The aim of the present study was to determine whether cholesterol efflux capacity (CEC), mediated by serum transporters (ABCA1 and ABCG1) and passive diffusion (PD), impacts on clinical outcome of advanced non-small cell lung cancer (NSCLC) and metastatic renal cell carcinoma (mRCC) pts treated with ICIs. MATERIAL AND METHODS We retrospectively enrolled advanced NSCLC and mRCC pts consecutively treated with ICIs between October 2013 and October 2018. CEC and cholesterol loading capacity (CLC) were assessed by well-established specific cell models. As primary endpoint, CEC, PD and CLC were correlated with overall survival (OS) while the effects of these parameters on progression-free survival (PFS) and clinical benefit (CB), defined as complete/partial response or stable disease, represented secondary endpoints. RESULTS NSCLC accounted for 94.2% of 70 enrolled cases, and serum sample suitable for CEC and PD determination was available in 68. Blood cholesterol and serum ABCA1, ABCG1, PD and CLC were associated with outcomes (OS, PFS and CB) at univariate analysis. At the multivariate analysis, only PD confirmed its positive prognostic value in terms of OS, PFS and CB. CONCLUSION The favorable impact of cholesterol PD on clinical outcome might reflect its main conformation in mature HDL particles which potentially shape an inflamed context, ultimately promoting ICI efficacy. Further prospective studies are needed to support our findings and uncover targetable pathways.
Collapse
Affiliation(s)
- Fabiana Perrone
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - Elda Favari
- Food and Drug Department, University of Parma, Parma, Italy
| | - Giuseppe Maglietta
- Clinical & Epidemiological Research Unit, University Hospital of Parma, Parma, Italy
| | - Michela Verzè
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Monica Pluchino
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Roberto Sabato
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Annalisa Ronca
- Food and Drug Department, University of Parma, Parma, Italy
| | | | - Alessio Cortellini
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Federica Pecci
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Ancona, Italy
| | - Luca Cantini
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti, Ancona, Italy
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
8
|
You Y, Fan H, Zhang S, Hu S, Tang J, Chen C, Wen W, Wang C, Cheng Y, Zhou M, Feng Z, Tan T, Qi G, Zhao W, Zhang X, Wang M, Dai L. Reduced plasma cholesterol in Plasmodium falciparum infection: A meta-analysis. J Infect 2023; 87:e19-e21. [PMID: 37172785 DOI: 10.1016/j.jinf.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/25/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023]
Affiliation(s)
- Yao You
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Hua Fan
- School of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan University of Science and Technology, Luoyang 471003, Henan, China
| | - Shenghui Zhang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Siqi Hu
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Jiake Tang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Chen Chen
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Wen Wen
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Chunyi Wang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Yongran Cheng
- School of Public Health, Hangzhou Medical College, Hangzhou 311300, China
| | - Mengyun Zhou
- Department of Molecular & Cellular Physiology, Shinshu University School of Medicine, 3900803, Japan
| | - Zhanhui Feng
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tao Tan
- Faculty of Applied Science, Macao Polytechnic University, 999078, Macao Special Administrative Region of China
| | - Guanming Qi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, Boston, MA 02111, USA
| | - Wenbin Zhao
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China
| | - Xingwei Zhang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China.
| | - Mingwei Wang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China.
| | - Lili Dai
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou 310015, China; Hangzhou Lin'an Fourth People's Hospital, Hangzhou 311321, China.
| |
Collapse
|
9
|
Huang D, Zheng S, Huang F, Chen J, Zhang Y, Chen Y, Li B. Prognostic nomograms integrating preoperative serum lipid derivative and systemic inflammatory marker of patients with non-metastatic colorectal cancer undergoing curative resection. Front Oncol 2023; 13:1100820. [PMID: 36969019 PMCID: PMC10034181 DOI: 10.3389/fonc.2023.1100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
BackgroundLipid metabolism and cancer-related inflammation are closely related to the progression and prognosis of colorectal cancer (CRC). Therefore, this study aims to establish novel nomograms based on the combined detection of preoperative blood lipids and systemic inflammatory indicators to predict the overall survival (OS) and cancer-specific survival (CCS) of CRC patients.MethodsA total of 523 patients with stage I-III CRC in our institute were collected from 2014 to 2018. The independent predictors for OS and CCS were determined by forward stepwise Cox regression for the establishment of prognostic models. The superiorities of different models were compared by concordance index (C-index), Akaike information criterion (AIC) and integrated discrimination improvement analysis. The performance of the nomograms based on the optimal models was measured by the plotting time-dependent receiver operating characteristic curves, calibration curves, and decision curves, and compared with the tumor-node-metastasis (TNM) staging system. The cohort was categorized into low-risk, medium-risk and high-risk groups according to the risk points of the nomogram, and analyzed using Kaplan–Meier curves and log-rank test.ResultsPreoperative TG/HDL-C ratio (THR) ≥ 1.93 and prognostic nutritional index (PNI) ≥ 42.55 were independently associated with favorable outcomes in CRC patients. Six (pT stage, pN stage, histological subtype, perineural invasion, THR and PNI) and seven (pT stage, pN stage, histological subtype, perineural invasion, gross appearance, THR and PNI) variables were chosen to develop the optimal models and construct nomograms for the prediction of OS and CCS. The models had lower AIC and larger C-indexes than other models lacking either or both of THR and PNI, and improved those integrated discrimination ability significantly. The nomograms showed better discrimination ability, calibration ability and clinical effectiveness than TNM system in predicting OS and CCS, and these results were reproducible in the validation cohort. The three risk stratifications based on the nomograms presented significant discrepancies in prognosis.ConclusionPreoperative THR and PNI have distinct prognostic value in stage I-III CRC patients. The nomograms incorporated the two indexes provide an intuitive and reliable approach for predicting the prognosis and optimizing individualized therapy of non-metastatic CRC patients, which may be a complement to the TNM staging system.
Collapse
Affiliation(s)
- Dimei Huang
- Department of General Internal Medicine, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Shaochu Zheng
- Department of Haematology/Oncology and Paediatric Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Fang Huang
- Department of General Internal Medicine, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Jingyu Chen
- Department of General Internal Medicine, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Yuexiang Zhang
- Department of Oncology, Affiliated Changzhi People’s Hospital of Changzhi Medical College, Changzhi, China
| | - Yusha Chen
- Department of Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Bixun Li
- Department of General Internal Medicine, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Bixun Li,
| |
Collapse
|
10
|
Ronca A, Pellegrini N, Pagliai G, Dinu M, Manfredini M, Incerti M, Favari E, Sofi F. Effects of a dietary intervention with Mediterranean vs lacto-ovo vegetarian diets on HDL function: Results from the CARDIVEG study. Nutr Metab Cardiovasc Dis 2023; 33:651-658. [PMID: 36642608 DOI: 10.1016/j.numecd.2022.11.012] [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: 04/14/2022] [Revised: 09/18/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND AIM HDL-cholesterol efflux capacity (CEC) has been shown to be a better cardiovascular (CVD) risk marker than serum HDL concentration. Several foods and nutrients have been shown to improve HDL functions, however no effective dietetic nor pharmacological strategy is available to increase CEC. This study aims to evaluate the possible effect of Mediterranean diet (MD) and lacto-ovo-vegetarian diet (VD) on HDL function in a group of clinically healthy subjects at low-to-moderate CVD risk. METHODS AND RESULTS Thirty apparently healthy subjects with a low-to-moderate cardiovascular risk profile (21 F; mean age: 51.3 ± 9.7 years) were randomly assigned to a 3-month MD or VD diet and then crossed. Participants on VD showed a reduction in total HDL CEC by 8.99% (p < 0.001) as well as a reduction in ABCA1 mediated-CEC by 18.62% (p < 0.001) compared to participants on MD. Regarding CEC mediated by aqueous diffusion, no significant changes were observed after treatment with either diet. Finally, a significant positive association between CEC mediated by the ABCA1 transporter and adiponectin was found (r = 0.462; p = 0.010). CONCLUSION The results of this study suggest that HDL activity in promoting cholesterol efflux and thereby reducing the concentration of pro-atherogenic lipoproteins was more effective in participants undergoing MD than VD. Based on these findings, the MD could be considered a better therapeutic strategy for cardiovascular prevention than VD. CLINICAL TRIAL REGISTRATION URL http://www. CLINICALTRIALS gov. Unique identifier: NCT02641834.
Collapse
Affiliation(s)
- Annalisa Ronca
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Nicoletta Pellegrini
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Giuditta Pagliai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Monica Dinu
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Manfredini
- Department of Chemistry, Life Science, And Environmental Sustainability, University of Parma, Parma, Italy
| | - Matteo Incerti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy.
| | - Francesco Sofi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
11
|
Papagiannis A, Gkolfinopoulou C, Tziomalos K, Dedemadi AG, Polychronopoulos G, Milonas D, Savopoulos C, Hatzitolios AI, Chroni A. HDL cholesterol efflux capacity and phospholipid content are associated with the severity of acute ischemic stroke and predict its outcome. Clin Chim Acta 2023; 540:117229. [PMID: 36657609 DOI: 10.1016/j.cca.2023.117229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND/AIMS Impaired high-density lipoprotein (HDL) function and composition are more strongly related to cardiovascular morbidity than HDL concentration. However, it is unclear whether HDL function and composition predict ischemic stroke severity and outcome. We aimed to evaluate these associations. METHODS We prospectively studied 199 consecutive patients who were admitted with acute ischemic stroke. The severity of stroke was evaluated at admission with the National Institutes of Health Stroke Scale (NIHSS). Severe stroke was defined as NIHSS ≥ 5. The outcome was assessed with dependency at discharge (modified Rankin scale 2-5) and in-hospital mortality. Cholesterol efflux capacity (CEC), phospholipid levels, lecithin:cholesterol acyl transferase (LCAT)-phospholipase activity, paraoxonase-1 (PON1)-arylesterase activity and serum amyloid A1 (SAA1) content of HDL were measured. RESULTS CEC, phospholipid levels and LCAT-phospholipase activity of HDL were lower and SAA1 content of HDL was higher in patients with severe stroke. Patients who were dependent at discharge had lower CEC, PON1-arylesterase activity, phospholipid content and LCAT-phospholipase activity of HDL and higher HDL-SAA1 content. Independent predictors of dependency at discharge were the NIHSS at admission (RR 2.60, 95% CI 1.39-4.87), lipid-lowering treatment (RR 0.17, 95% CI 0.01-0.75), HDL-CEC (RR 0.21, 95% CI 0.05-0.87) and HDL-associated PON1-arylesterase activity (RR 0.95, 95% CI 0.91-0.99). In patients who died during hospitalization, phospholipids, LCAT-phospholipase and PON1-arylesterase activities of HDL were lower. CONCLUSIONS Changes in CEC and composition of HDL appear to be associated with the severity and outcome of acute ischemic stroke and could represent biomarkers that may inform risk stratification and management strategies in these patients.
Collapse
Affiliation(s)
- Achilleas Papagiannis
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Konstantinos Tziomalos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece.
| | - Anastasia-Georgia Dedemadi
- Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Georgios Polychronopoulos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Dimitrios Milonas
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Christos Savopoulos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Apostolos I Hatzitolios
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece.
| |
Collapse
|
12
|
Ronda N, Zimetti F, Adorni MP, Palumbo M, Karpouzas GA, Bernini F. Role of Lipoprotein Levels and Function in Atherosclerosis Associated with Autoimmune Rheumatic Diseases. Rheum Dis Clin North Am 2023; 49:151-163. [PMID: 36424022 DOI: 10.1016/j.rdc.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Immune and inflammatory mediators in autoimmune rheumatic diseases induce modification in the activity of enzymes pivotal for lipid metabolism and promote a proatherogenic serum lipid profile. However, disturbances in low- and high-density lipoprotein composition and increased lipid oxidation also occur. Therefore, lipoprotein dysfunction causes intracellular cholesterol accumulation in macrophages, smooth muscle cells, and platelets. Overall, both plaque progression and acute cardiovascular events are promoted. Single rheumatic diseases may present a particular pattern of lipid disturbances so that standard methods to evaluate cardiovascular risk may not be accurate enough. In general, antirheumatic drugs positively affect lipid metabolism in these patients.
Collapse
Affiliation(s)
- Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy.
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, Via Volturno 39/F, Parma 43125, Italy
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy
| | - George A Karpouzas
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute, Torrance, CA, USA
| | - Franco Bernini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, Parma 43124, Italy
| |
Collapse
|
13
|
A Transient Inflammatory Response Induced by Lipopolysaccharide Infusion Lowers Markers of Endogenous Cholesterol and Bile Acid Synthesis in Healthy Normocholesterolemic Young Men. Biomedicines 2023; 11:biomedicines11010126. [PMID: 36672634 PMCID: PMC9855383 DOI: 10.3390/biomedicines11010126] [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: 12/05/2022] [Revised: 12/27/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Inflammation is associated with changes in plasma lipids, lipoproteins, and cholesterol efflux capacity (CEC). It is unknown if the changes in lipids and lipoproteins during inflammation are related to changes in cholesterol absorption, synthesis, and bile acid synthesis. We, therefore, examined the effects of acute lipopolysaccharide (LPS)-induced transient systemic inflammation on lipids, lipoproteins, CEC, and markers of cholesterol metabolism. We also evaluated whether markers for cholesterol metabolism at baseline predict the intensity of the inflammatory response. Eight healthy young subjects received LPS infusion, and blood was sampled for the following 24 h. In addition to lipids, lipoproteins, and CEC, we also measured markers for cholesterol absorption and synthesis, bile acid synthesis, and inflammation. Compared with baseline, plasma total cholesterol, low-density lipoprotein cholesterol, and CEC decreased, while triglycerides increased in the 24 h following LPS infusion. TC-standardized levels of cholesterol synthesis markers (lathosterol, lanosterol, and desmosterol) and a bile acid synthesis marker (7α-OH-cholesterol) also decreased, with no changes in cholesterol absorption markers (campesterol, sitosterol, and cholestanol). Baseline TC-standardized levels of desmosterol and 7α-OH-cholesterol were positively correlated with concentrations of various inflammatory markers. Changes in TC-standardized desmosterol and 7α-OH-cholesterol were negatively correlated with concentrations of inflammatory markers. LPS infusion reduced endogenous cholesterol synthesis and bile acid synthesis in healthy young men.
Collapse
|
14
|
Effects of Antirheumatic Treatment on Cell Cholesterol Efflux and Loading Capacity of Serum Lipoproteins in Spondylarthropathies. J Clin Med 2022; 11:jcm11247330. [PMID: 36555946 PMCID: PMC9780876 DOI: 10.3390/jcm11247330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Spondyloarthropathies (SpA) are associated with increased cardiovascular risk. Among possible mechanisms is the dysfunction of serum lipoproteins in regulating cell cholesterol homeostasis. Cholesterol efflux capacity (CEC)-the atheroprotective ability of HDL (high density lipoproteins) to accept cholesterol from macrophages-might predict cardiovascular disease independently of HDL-cholesterol levels. We aimed at evaluating modifications of CEC and of the atherogenic cholesterol loading capacity (CLC) of serum lipoproteins in psoriatic arthritis (PsA) and ankylosing spondylitis (AS) following anti-rheumatic treatment. A total of 62 SpA patients (37 PsA and 25 AS) were evaluated before and after treatment with tumor necrosis factor inhibitor and/or methotrexate. CEC and CLC were measured by radioisotopic and fluorometric techniques, respectively. Endothelial function was assessed by finger plethysmography (Endopat). In the whole SpA group, total and HDL-cholesterol increased after treatment, while lipoprotein(a) decreased and CLC was unchanged. Treatment was associated with increased Scavenger Receptor class B type I (SR-BI)-mediated CEC in the AS group. SR-BI- and ABCG1-mediated CEC were negatively associated with inflammatory parameters and positively related to coffee consumption. SR-BI CEC and CLC were positively and negatively associated with endothelial function, respectively. Our pilot study suggests that anti-rheumatic treatment is associated with favorable modulation of lipoprotein quality and function in SpA, particularly in AS, in spite of the induced increase in total cholesterol levels. If confirmed in a larger population, this might represent an atheroprotective benefit beyond what is reflected by conventional serum lipid profile.
Collapse
|
15
|
Lalosevic M, Kotur-Stevuljevic J, Vekic J, Rizzo M, Kosanovic T, Blagojevic IP, Zeljkovic A, Jeremic D, Mihajlovic M, Petkovic A, Hajdarpasic L, Djordjevic M, Dobrilovic V, Erceg S, Vujcic S, Marjanovic J, Jovanovic JM, Saponjski J, Bogavac-Stanojevic N. Alteration in Redox Status and Lipoprotein Profile in COVID-19 Patients with Mild, Moderate, and Severe Pneumonia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8067857. [PMID: 36420478 PMCID: PMC9678464 DOI: 10.1155/2022/8067857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/24/2022] [Accepted: 11/05/2022] [Indexed: 08/23/2023]
Abstract
Background Metabolic alterations, particularly disorders of lipoprotein metabolism in COVID-19, may affect the course and outcome of the disease. This study aims at evaluating the lipoprotein profile and redox status in SARS-CoV-2 infected patients with different pneumonia severity and their association with lethal outcomes. Methods The prospective cohort study was performed on 98 COVID-19 patients with mild, moderate, and severe pneumonia. Lipid and inflammatory parameters, lipoprotein subclasses, and redox status biomarkers were determined at the study entry and after one week. Results Compared to patients with mild and moderate pneumonia, severely ill patients had higher oxidised low-density lipoprotein (oxLDL) and malondialdehyde levels and lower high-density lipoprotein cholesterol (HDL-C) concentrations and paraoxonase 1 activity. Reduction in the proportion of large HDL 2a subclasses with a concomitant increase in the proportion of smallest HDL 3c and small dense LDL (sdLDL) particles was observed in patients with severe disease during the time. However, these changes were reversed in the mild and moderate groups. The results showed a positive association between changes in oxLDL and total antioxidative status. However, prooxidants and antioxidants in plasma were lower in patients with lethal outcomes. Conclusions Increased levels of oxLDL and sdLDL particles may contribute to the severity of COVID-19. The role of oxidative stress should be clarified in further studies, mainly its association with lethal outcomes.
Collapse
Affiliation(s)
- Miodrag Lalosevic
- Radiology Department, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Jelena Kotur-Stevuljevic
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Jelena Vekic
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties, University of Palermo, Palermo, Italy
| | - Tijana Kosanovic
- Radiology Department, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Iva Perovic Blagojevic
- Department of Laboratory Diagnostic, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Aleksandra Zeljkovic
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Danilo Jeremic
- Orthopedics Department, Institute for Orthopedic Surgery “Banjica”, Belgrade, Serbia
| | - Marija Mihajlovic
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Aleksa Petkovic
- Department of Laboratory Diagnostic, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Lejla Hajdarpasic
- Radiology Department, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Marjana Djordjevic
- Radiology Department, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Violeta Dobrilovic
- Radiology Department, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Sanja Erceg
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Vujcic
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Jelena Marjanovic
- Department of Laboratory Diagnostic, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Jovana Milijic Jovanovic
- Department of Laboratory Diagnostic, University Hospital “Dr. Dragisa Misovic-Dedinje”, Belgrade, Serbia
| | - Jovica Saponjski
- Cardiology Clinic, University Clinical Centre of Serbia, Belgrade, Serbia
| | | |
Collapse
|
16
|
Adorni MP, Palumbo M, Marchi C, Zimetti F, Ossoli A, Turri M, Bernini F, Hollan I, Moláček J, Treska V, Ronda N. HDL metabolism and functions impacting on cell cholesterol homeostasis are specifically altered in patients with abdominal aortic aneurysm. Front Immunol 2022; 13:935241. [PMID: 36172376 PMCID: PMC9510680 DOI: 10.3389/fimmu.2022.935241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe etiopathogenesis of abdominal aortic aneurysm (AAA) is still unclarified, but vascular inflammation and matrix metalloproteases activation have a recognized role in AAA development and progression. Circulating lipoproteins are involved in tissue inflammation and repair, particularly through the regulation of intracellular cholesterol, whose excess is associated to cell damage and proinflammatory activation. We analyzed lipoprotein metabolism and function in AAA and in control vasculopathic patients, to highlight possible non-atherosclerosis-related, specific abnormalities.MethodsWe measured fluorometrically serum esterified/total cholesterol ratio, as an index of lecithin-cholesterol acyltransferase (LCAT) activity, and cholesteryl ester transfer protein (CETP) activity in patients referred to vascular surgery either for AAA (n=30) or stenotic aortic/peripheral atherosclerosis (n=21) having similar burden of cardiovascular risk factors and disease. We measured high-density lipoprotein (HDL)-cholesterol efflux capacity (CEC), through the ATP-binding cassette G1 (ABCG1) and A1 (ABCA1) pathways and serum cell cholesterol loading capacity (CLC), by radioisotopic and fluorimetric methods, respectively.ResultsWe found higher LCAT (+23%; p < 0.0001) and CETP (+49%; p < 0.0001) activity in AAA sera. HDL ABCG1-CEC was lower (−16%; p < 0.001) and ABCA1-CEC was higher (+31.7%; p < 0.0001) in AAA. Stratification suggests that smoking may partly contribute to these modifications. CEC and CETP activity correlated with CLC only in AAA.ConclusionsWe demonstrated that compared to patients with stenotic atherosclerosis, patients with AAA had altered HDL metabolism and functions involved in their anti-inflammatory and tissue repair activity, particularly through the ABCG1-related intracellular signaling. Clarifying the relevance of this mechanism for AAA evolution might help in developing new diagnostic parameters and therapeutic targets for the early management of this condition.
Collapse
Affiliation(s)
- Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, Via Volturno 39/F, Parma, Italy
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma, Italy
| | - Cinzia Marchi
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma, Italy
| | - Alice Ossoli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Marta Turri
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma, Italy
- *Correspondence: Franco Bernini,
| | - Ivana Hollan
- Lillehammer Hospital for Rheumatic Diseases, M. Grundtvigs veg 6, Lillehammer, Norway and Brigham and Women’s Hospital, Cardiology Division, Boston, United States
| | - Jiří Moláček
- Department of Vascular Surgery, Faculty of Medicine and University Hospital in Plzen, Charles University Ovocný trh 5 Prague 1, Plzen, Czechia
| | - Vladislav Treska
- Department of Vascular Surgery, Faculty of Medicine and University Hospital in Plzen, Charles University Ovocný trh 5 Prague 1, Plzen, Czechia
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma, Italy
| |
Collapse
|
17
|
Adorni MP, Biolo M, Zimetti F, Palumbo M, Ronda N, Scarinzi P, Simioni P, Lupo MG, Ferri N, Previato L, Bernini F, Zambon A. HDL Cholesterol Efflux and Serum Cholesterol Loading Capacity Alterations Associate to Macrophage Cholesterol Accumulation in FH Patients with Achilles Tendon Xanthoma. Int J Mol Sci 2022; 23:ijms23158255. [PMID: 35897824 PMCID: PMC9332368 DOI: 10.3390/ijms23158255] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 02/05/2023] Open
Abstract
Achilles tendon xanthoma (ATX) formation involves macrophage cholesterol accumulation within the tendon, similar to that occurring in atheroma. Macrophage cholesterol homeostasis depends on serum lipoprotein functions, namely the high-density lipoprotein (HDL) capacity to promote cell cholesterol efflux (cholesterol efflux capacity, CEC) and the serum cholesterol loading capacity (CLC). We explored the HDL-CEC and serum CLC, comparing 16 FH patients with ATX to 29 FH patients without ATX. HDL-CEC through the main efflux mechanisms mediated by the transporters ATP binding cassette G1 (ABCG1) and A1 (ABCA1) and the aqueous diffusion (AD) process was determined by a cell-based radioisotopic technique and serum CLC fluorimetrically. Between the two groups, no significant differences were found in terms of plasma lipid profile. A trend toward reduction of cholesterol efflux via AD and a significant increase in ABCA1-mediated HDL-CEC (+18.6%) was observed in ATX compared to no ATX patients. In ATX-presenting patients, ABCG1-mediated HDL-CEC was lower (−11%) and serum CLC was higher (+14%) compared to patients without ATX. Considering all the patients together, ABCG1 HDL-CEC and serum CLC correlated with ATX thickness inversely (p = 0.013) and directly (p < 0.0001), respectively. In conclusion, lipoprotein dysfunctions seem to be involved in ATX physiopathology and progression in FH patients.
Collapse
Affiliation(s)
- Maria Pia Adorni
- Unit of Neuroscience, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy;
| | - Marta Biolo
- Department of Medicine, University of Padua, 35128 Padua, Italy; (M.B.); (P.S.); (P.S.); (M.G.L.); (N.F.); (L.P.)
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (F.Z.); (M.P.); (N.R.)
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (F.Z.); (M.P.); (N.R.)
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (F.Z.); (M.P.); (N.R.)
| | - Paolo Scarinzi
- Department of Medicine, University of Padua, 35128 Padua, Italy; (M.B.); (P.S.); (P.S.); (M.G.L.); (N.F.); (L.P.)
| | - Paolo Simioni
- Department of Medicine, University of Padua, 35128 Padua, Italy; (M.B.); (P.S.); (P.S.); (M.G.L.); (N.F.); (L.P.)
| | - Maria Giovanna Lupo
- Department of Medicine, University of Padua, 35128 Padua, Italy; (M.B.); (P.S.); (P.S.); (M.G.L.); (N.F.); (L.P.)
| | - Nicola Ferri
- Department of Medicine, University of Padua, 35128 Padua, Italy; (M.B.); (P.S.); (P.S.); (M.G.L.); (N.F.); (L.P.)
| | - Lorenzo Previato
- Department of Medicine, University of Padua, 35128 Padua, Italy; (M.B.); (P.S.); (P.S.); (M.G.L.); (N.F.); (L.P.)
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (F.Z.); (M.P.); (N.R.)
- Correspondence: ; Tel.: +39-0521-905039
| | | |
Collapse
|
18
|
Palumbo M, Giammanco A, Purrello F, Pavanello C, Mombelli G, Di Pino A, Piro S, Cefalù AB, Calabresi L, Averna M, Bernini F, Zimetti F, Adorni MP, Scicali R. Effects of PCSK9 inhibitors on HDL cholesterol efflux and serum cholesterol loading capacity in familial hypercholesterolemia subjects: a multi-lipid-center real-world evaluation. Front Mol Biosci 2022; 9:925587. [PMID: 35928226 PMCID: PMC9343790 DOI: 10.3389/fmolb.2022.925587] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/27/2022] [Indexed: 01/03/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9), beyond regulating LDL cholesterol (LDL-c) plasma levels, exerts several pleiotropic effects by modulating lipid metabolism in extrahepatic cells such as macrophages. Macrophage cholesterol homeostasis depends on serum lipoprotein functions, including the HDL capacity to promote cell cholesterol efflux (CEC) and the serum capacity to promote cell cholesterol loading (CLC). The aim of this observational study was to investigate the effect of PCSK9 inhibitors (PCSK9-i) treatment on HDL-CEC and serum CLC in patients with familial hypercholesterolemia (FH). 31 genetically confirmed FH patients were recruited. Blood was collected and serum isolated at baseline and after 6 months of PCSK9-i treatment. HDL-CEC was evaluated through the main pathways with a radioisotopic cell-based assay. Serum CLC was assessed fluorimetrically in human THP-1 monocyte-derived macrophages. After treatment with PCSK9-i, total cholesterol and LDL-c significantly decreased (−41.6%, p < 0.0001 and −56.7%, p < 0.0001, respectively). Total HDL-CEC was not different between patients before and after treatment. Conversely, despite no changes in HDL-c levels between the groups, ABCG1 HDL-CEC significantly increased after treatment (+22.2%, p < 0.0001) as well as HDL-CEC by aqueous diffusion (+7.8%, p = 0.0008). Only a trend towards reduction of ABCA1 HDL-CEC was observed after treatment. PCSK9-i significantly decreased serum CLC (−6.6%, p = 0.0272). This effect was only partly related to the reduction of LDL-c levels. In conclusion, PCSK9-i treatment significantly increased HDL-CEC through ABCG1 and aqueous diffusion pathways and reduced the serum CLC in FH patients. The favorable effect of PCSK9-i on functional lipid profile could contribute to the cardiovascular benefit of these drugs in FH patients.
Collapse
Affiliation(s)
| | - Antonina Giammanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE)—University of Palermo, Palermo, Italy
| | - Francesco Purrello
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
- Centro Dislipidemie, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Giuliana Mombelli
- Centro Dislipidemie, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Antonino Di Pino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Angelo Baldassare Cefalù
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE)—University of Palermo, Palermo, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE)—University of Palermo, Palermo, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parma, Italy
- *Correspondence: Francesca Zimetti,
| | - Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, Parma, Italy
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
19
|
Zhang J, Dai W, Chen Y. Editorial: The Roles of Lipids in Immunometabolism: The Crosstalk Between Lipid Metabolisms and Inflammation. Front Cardiovasc Med 2022; 9:938535. [PMID: 35811702 PMCID: PMC9257247 DOI: 10.3389/fcvm.2022.938535] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Jue Zhang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Wen Dai
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Yiliang Chen
| |
Collapse
|
20
|
Ong KL, Cochran BBiotech BJ, Manandhar B, Thomas S, Rye KA. HDL maturation and remodelling. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159119. [PMID: 35121104 DOI: 10.1016/j.bbalip.2022.159119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 11/29/2022]
Abstract
Cholesterol in the circulation is mostly transported in an esterified form as a component of lipoproteins. The majority of these cholesteryl esters are produced in nascent, discoidal high density lipoproteins (HDLs) by the enzyme, lecithin:cholesterol acyltransferase (LCAT). Discoidal HDLs are discrete populations of particles that consist of a phospholipid bilayer, the hydrophobic acyl chains of which are shielded from the aqueous environment by apolipoproteins that also confer water solubility on the particles. The progressive LCAT-mediated accumulation of cholesteryl esters in discoidal HDLs generates the spherical HDLs that predominate in normal human plasma. Spherical HDLs contain a core of water insoluble, neutral lipids (cholesteryl esters and triglycerides) that is surrounded by a surface monolayer of phospholipids with which apolipoproteins associate. Although spherical HDLs all have the same basic structure, they are extremely diverse in size, composition, and function. This review is concerned with how the biogenesis of discoidal and spherical HDLs is regulated and the mechanistic basis of their size and compositional heterogeneity. Current understanding of the impact of this heterogeneity on the therapeutic potential of HDLs of varying size and composition is also addressed in the context of several disease states.
Collapse
Affiliation(s)
- Kwok-Leung Ong
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Blake J Cochran BBiotech
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Bikash Manandhar
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Shane Thomas
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, New South Wales, Australia.
| |
Collapse
|
21
|
Papotti B, Macchi C, Favero C, Iodice S, Adorni MP, Zimetti F, Corsini A, Aliberti S, Blasi F, Carugo S, Bollati V, Vicenzi M, Ruscica M. HDL in COVID-19 Patients: Evidence from an Italian Cross-Sectional Study. J Clin Med 2021; 10:jcm10245955. [PMID: 34945250 PMCID: PMC8708284 DOI: 10.3390/jcm10245955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 02/07/2023] Open
Abstract
A number of studies have highlighted important alterations of the lipid profile in COVID-19 patients. Besides the well-known atheroprotective function, HDL displays anti-inflammatory, anti-oxidative, and anti-infectious properties. The aim of this retrospective study was to assess the HDL anti-inflammatory and antioxidant features, by evaluation of HDL-associated Serum amyloid A (SAA) enrichment and HDL-paraoxonase 1 (PON-1) activity, in a cohort of COVID-19 patients hospitalized at the Cardiorespiratory COVID-19 Unit of Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico of Milan. COVID-19 patients reached very low levels of HDL-c (mean ± SD: 27.1 ± 9.7 mg/dL) with a marked rise in TG (mean ± SD: 165.9 ± 62.5 mg/dL). Compared to matched-controls, SAA levels were significantly raised in COVID-19 patients at admission. There were no significant differences in the SAA amount between 83 alive and 22 dead patients for all-cause in-hospital mortality. Similar findings were reached in the case of PON-1 activity, with no differences between alive and dead patients for all-cause in-hospital mortality. In conclusion, although not related to the prediction of in-hospital mortality, reduction in HDL-c and the enrichment of SAA in HDL are a mirror of SARS-CoV-2 positivity even at the very early stages of the infection.
Collapse
Affiliation(s)
- Bianca Papotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (B.P.); (F.Z.)
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy; (C.M.); (A.C.)
| | - Chiara Favero
- EPIGET Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20100 Milan, Italy; (C.F.); (S.I.); (V.B.)
| | - Simona Iodice
- EPIGET Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20100 Milan, Italy; (C.F.); (S.I.); (V.B.)
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy;
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (B.P.); (F.Z.)
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy; (C.M.); (A.C.)
- IRCCS Multimedica, 20099 Sesto San Giovanni, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, 20100 Milan, Italy;
| | - Francesco Blasi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Adult Center, 20100 Milan, Italy;
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20100 Milan, Italy
| | - Stefano Carugo
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Cardiovascular Disease Unit, Internal Medicine Department, 20100 Milan, Italy;
- Dyspnea Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20100 Milan, Italy
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20100 Milan, Italy; (C.F.); (S.I.); (V.B.)
| | - Marco Vicenzi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Cardiovascular Disease Unit, Internal Medicine Department, 20100 Milan, Italy;
- Dyspnea Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20100 Milan, Italy
- Correspondence: (M.V.); (M.R.)
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy; (C.M.); (A.C.)
- Correspondence: (M.V.); (M.R.)
| |
Collapse
|
22
|
Garcia AR, Finch C, Gatz M, Kraft T, Eid Rodriguez D, Cummings D, Charifson M, Buetow K, Beheim BA, Allayee H, Thomas GS, Stieglitz J, Gurven MD, Kaplan H, Trumble BC. APOE4 is associated with elevated blood lipids and lower levels of innate immune biomarkers in a tropical Amerindian subsistence population. eLife 2021; 10:68231. [PMID: 34586066 PMCID: PMC8480980 DOI: 10.7554/elife.68231] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
In post-industrial settings, apolipoprotein E4 (APOE4) is associated with increased cardiovascular and neurological disease risk. However, the majority of human evolutionary history occurred in environments with higher pathogenic diversity and low cardiovascular risk. We hypothesize that in high-pathogen and energy-limited contexts, the APOE4 allele confers benefits by reducing innate inflammation when uninfected, while maintaining higher lipid levels that buffer costs of immune activation during infection. Among Tsimane forager-farmers of Bolivia (N = 1266, 50% female), APOE4 is associated with 30% lower C-reactive protein, and higher total cholesterol and oxidized LDL. Blood lipids were either not associated, or negatively associated with inflammatory biomarkers, except for associations of oxidized LDL and inflammation which were limited to obese adults. Further, APOE4 carriers maintain higher levels of total and LDL cholesterol at low body mass indices (BMIs). These results suggest that the relationship between APOE4 and lipids may be beneficial for pathogen-driven immune responses and unlikely to increase cardiovascular risk in an active subsistence population.
Collapse
Affiliation(s)
- Angela R Garcia
- Center for Evolution and Medicine, Arizona State University, Tempe, United States.,Department of Anthropology, Emory University, Atlanta, United States
| | - Caleb Finch
- Leonard Davis School of Gerontology, Dornsife College, University of Southern California, Los Angeles, Los Angeles, United States
| | - Margaret Gatz
- Center for Economic and Social Research, University of Southern California, Los Angeles, Los Angeles, United States
| | - Thomas Kraft
- Department of Anthropology, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Daniel Cummings
- Institute for Economics and Society, Chapman University, Orange, United States
| | - Mia Charifson
- Vilcek Institute of Graduate Biomedical Sciences, New York University, New York, United States
| | - Kenneth Buetow
- Center for Evolution and Medicine, Arizona State University, Tempe, United States.,School of Life Sciences, Arizona State University, Tempe, United States
| | - Bret A Beheim
- Department of Human Behavior, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Hooman Allayee
- Department of Preventive Medicine and Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Irvine, Irvine, United States.,Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Irvine, Irvine, United States
| | - Gregory S Thomas
- Long Beach Memorial, Long Beach and University of California Irvine, Irvine, United States
| | - Jonathan Stieglitz
- Institute for Advanced Study in Toulouse, Universite Toulouse, Toulouse, France
| | - Michael D Gurven
- Department of Anthropology, University of California, Santa Barbara, Santa Barbara, United States
| | - Hillard Kaplan
- Institute for Economics and Society, Chapman University, Orange, United States
| | - Benjamin C Trumble
- School of Human Evolution and Social Change, Arizona State University, Tempe, United States
| |
Collapse
|
23
|
Kocyigit D, Zimetti F, Gurses KM, Zanotti I, Marchi C, Ståhlman M, Borén J, Canpinar H, Soyal MF, Guc D, Hazirolan T, Ozer N, Tokgozoglu L. Cholesterol efflux promoting function of high-density lipoproteins in calcific aortic valve stenosis. ATHEROSCLEROSIS PLUS 2021; 44:18-24. [PMID: 36644669 PMCID: PMC9833266 DOI: 10.1016/j.athplu.2021.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 01/18/2023]
Abstract
Background and aims Cholesterol efflux capacity is a functional property of high-density lipoproteins (HDL) reflecting the efficiency of the atheroprotective reverse cholesterol transport process in humans. Its relationship with calcific aortic valve stenosis (CAVS) has not been fully assessed yet. Methods We evaluated HDL-CEC in a patient population with varying degrees of aortic valvular calcific disease, assessed using echocardiography and cardiac computed tomography. Measurement of biomarkers that reflect osteogenic and tissue remodeling, along with dietary and gut microbiota-derived metabolites were performed. Results Patients with moderate-severe CAVS had significantly lower HDL-CEC compared to both control and aortic sclerosis subjects (mean: 6.09%, 7.32% and 7.26%, respectively). HDL-CEC displayed negative correlations with peak aortic jet velocity and aortic valve calcium score, indexes of CAVS severity (ρ = -0.298, p = 0.002 and ρ = -0.358, p = 0.005, respectively). In multivariable regression model, HDL-CEC had independent association with aortic valve calcium score (B: -0.053, SE: 0.014, p < 0.001), GFR (B: -0.034, SE: 0.012, p = 0.007), as well as with levels of total cholesterol (B: 0.018, SE: 0.005, p = 0.002). Conclusion These results indicate an impairment of HDL-CEC in moderate-severe CAVS and may contribute to identify potential novel targets for CAVS management.
Collapse
Affiliation(s)
- Duygu Kocyigit
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
- Corresponding author. Department of Cardiology, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey.
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parma, Italy
- Corresponding author.
| | - Kadri M. Gurses
- Department of Basic Medical Sciences, Adnan Menderes University Faculty of Medicine, Aydin, Turkey
| | - Ilaria Zanotti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Cinzia Marchi
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine, University of Gothenburg Institute of Medicine, Göteborg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg Institute of Medicine, Göteborg, Sweden
| | - Hande Canpinar
- Department of Basic Oncology, Hacettepe University Institute of Oncology, Ankara, Turkey
| | - Mehmet F.T. Soyal
- Department of Cardiovascular Surgery, Medicana International Ankara Hospital, Ankara, Turkey
| | - Dicle Guc
- Department of Basic Oncology, Hacettepe University Institute of Oncology, Ankara, Turkey
| | - Tuncay Hazirolan
- Department of Radiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Necla Ozer
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Lale Tokgozoglu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
24
|
Morris G, Berk M, Walder K, O'Neil A, Maes M, Puri BK. The lipid paradox in neuroprogressive disorders: Causes and consequences. Neurosci Biobehav Rev 2021; 128:35-57. [PMID: 34118292 DOI: 10.1016/j.neubiorev.2021.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 04/27/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023]
Abstract
Chronic systemic inflammation is associated with an increased risk of cardiovascular disease in an environment of low low-density lipoprotein (LDL) and low total cholesterol and with the pathophysiology of neuroprogressive disorders. The causes and consequences of this lipid paradox are explored. Circulating activated neutrophils can release inflammatory molecules such as myeloperoxidase and the pro-inflammatory cytokines interleukin-1 beta, interleukin-6 and tumour necrosis factor-alpha. Since activated neutrophils are associated with atherosclerosis and cardiovascular disease and with major depressive disorder, bipolar disorder and schizophrenia, it seems reasonable to hypothesise that the inflammatory molecules released by them may act as mediators of the link between systemic inflammation and the development of atherosclerosis in neuroprogressive disorders. This hypothesis is tested by considering the association at a molecular level of systemic inflammation with increased LDL oxidation; increased small dense LDL levels; increased lipoprotein (a) concentration; secretory phospholipase A2 activation; cytosolic phospholipase A2 activation; increased platelet activation; decreased apolipoprotein A1 levels and function; decreased paroxonase-1 activity; hyperhomocysteinaemia; and metabolic endotoxaemia. These molecular mechanisms suggest potential therapeutic targets.
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
| | - Michael Berk
- 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; 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
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - 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
| | | |
Collapse
|
25
|
Karpouzas GA, Bui VL, Ronda N, Hollan I, Ormseth SR. Biologics and atherosclerotic cardiovascular risk in rheumatoid arthritis: a review of evidence and mechanistic insights. Expert Rev Clin Immunol 2021; 17:355-374. [PMID: 33673792 DOI: 10.1080/1744666x.2021.1899809] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Cardiovascular disease is a leading comorbidity in rheumatoid arthritis. Timely introduction of biologic therapies in a treat-to-target approach has optimized disease-related outcomes and attenuated accrual of comorbidities, including cardiovascular risk.Areas covered: A literature search in MEDLINE (via PubMed) was performed between January 2009 and November 2020. This manuscript explores recent developments in atherosclerotic cardiovascular risk in RA compared with non-RA individuals; it synopsizes differences in vascular function and inflammation, prevalence, burden, vulnerability, and progression of atherosclerotic plaque and their underlying cellular and molecular mechanisms. Finally, it reviews the recent literature on cardioprotective benefits of biologics and draws mechanistic links with inhibition of new plaque formation, stabilization of high-risk lesions and improvement in endothelial function, arterial stiffness, lipid metabolism, and traditional cardiac risk factors.Expert opinion: Increasing evidence points to a solid cardioprotective influence of earlier, longer, and ongoing use of biologic treatments in RA. Nevertheless, the precise mechanistic effects of plaque progression and remodeling, vascular stiffness, endothelial dysfunction, lipid metabolism, and traditional cardiac risk factors are less rigorously characterized.
Collapse
Affiliation(s)
- George A Karpouzas
- Division of Rheumatology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Viet L Bui
- Division of Rheumatology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Ivana Hollan
- The Norwegian University of Science and Technology, Gjøvik, Norway.,Beitostølen Sport and Health Centre, Beitostølen, Norway
| | - Sarah R Ormseth
- Division of Rheumatology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
26
|
Adorni MP, Ronda N, Bernini F, Zimetti F. High Density Lipoprotein Cholesterol Efflux Capacity and Atherosclerosis in Cardiovascular Disease: Pathophysiological Aspects and Pharmacological Perspectives. Cells 2021; 10:cells10030574. [PMID: 33807918 PMCID: PMC8002038 DOI: 10.3390/cells10030574] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Over the years, the relationship between high-density lipoprotein (HDL) and atherosclerosis, initially highlighted by the Framingham study, has been revealed to be extremely complex, due to the multiple HDL functions involved in atheroprotection. Among them, HDL cholesterol efflux capacity (CEC), the ability of HDL to promote cell cholesterol efflux from cells, has emerged as a better predictor of cardiovascular (CV) risk compared to merely plasma HDL-cholesterol (HDL-C) levels. HDL CEC is impaired in many genetic and pathological conditions associated to high CV risk such as dyslipidemia, chronic kidney disease, diabetes, inflammatory and autoimmune diseases, endocrine disorders, etc. The present review describes the current knowledge on HDL CEC modifications in these conditions, focusing on the most recent human studies and on genetic and pathophysiologic aspects. In addition, the most relevant strategies possibly modulating HDL CEC, including lifestyle modifications, as well as nutraceutical and pharmacological interventions, will be discussed. The objective of this review is to help understanding whether, from the current evidence, HDL CEC may be considered as a valid biomarker of CV risk and a potential pharmacological target for novel therapeutic approaches.
Collapse
Affiliation(s)
- Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy;
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (N.R.); (F.Z.)
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (N.R.); (F.Z.)
- Correspondence:
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (N.R.); (F.Z.)
| |
Collapse
|
27
|
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: 30] [Impact Index Per Article: 7.5] [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
|
28
|
Marked Changes in Serum Amyloid A Distribution and High-Density Lipoprotein Structure during Acute Inflammation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9241259. [PMID: 33575357 PMCID: PMC7861920 DOI: 10.1155/2021/9241259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022]
Abstract
High-density lipoprotein- (HDL-) cholesterol measurements are generally used in the diagnosis of cardiovascular diseases. However, HDL is a complicated heterogeneous lipoprotein, and furthermore, it can be converted into dysfunctional forms during pathological conditions including inflammation. Therefore, qualitative analysis of pathophysiologically diversified HDL forms is important. A recent study demonstrated that serum amyloid A (SAA) can remodel HDL and induce atherosclerosis not only over long periods of time, such as during chronic inflammation, but also over shorter periods. However, few studies have investigated rapid HDL remodeling. In this study, we analyzed HDL samples from patients undergoing orthopedic surgery inducing acute inflammation. We enrolled 13 otherwise healthy patients who underwent orthopedic surgery. Plasma samples were obtained on preoperative day and postoperative days (POD) 1-7. SAA, apolipoprotein A-I (apoA-I), and apolipoprotein A-II (apoA-II) levels in the isolated HDL were determined. HDL particle size, surface charge, and SAA and apoA-I distributions were also analyzed. In every patient, plasma SAA levels peaked on POD3. Consistently, the HDL apoA-I : apoA-II ratio markedly decreased at this timepoint. Native-polyacrylamide gel electrophoresis and high-performance liquid chromatography revealed the loss of small HDL particles during acute inflammation. Furthermore, HDL had a decreased negative surface charge on POD3 compared to the other timepoints. All changes observed were SAA-dependent. SAA-dependent rapid changes in HDL size and surface charge were observed after orthopedic surgery. These changes might affect the atheroprotective functions of HDL, and its analysis can be available for the qualitative HDL assessment.
Collapse
|
29
|
Zimetti F, Adorni MP, Marsillach J, Marchi C, Trentini A, Valacchi G, Cervellati C. Connection between the Altered HDL Antioxidant and Anti-Inflammatory Properties and the Risk to Develop Alzheimer's Disease: A Narrative Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6695796. [PMID: 33505588 PMCID: PMC7811424 DOI: 10.1155/2021/6695796] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023]
Abstract
The protein composition of high-density lipoprotein (HDL) is extremely fluid. The quantity and quality of protein constituents drive the multiple biological functions of these lipoproteins, which include the ability to contrast atherogenesis, sustained inflammation, and toxic effects of reactive species. Several diseases where inflammation and oxidative stress participate in the pathogenetic process are characterized by perturbation in the HDL proteome. This change inevitably affects the functionality of the lipoprotein. An enlightening example in this frame comes from the literature on Alzheimer's disease (AD). Growing lines of epidemiological evidence suggest that loss of HDL-associated proteins, such as lipoprotein phospholipase A2 (Lp-PLA2), glutathione peroxidase-3 (GPx-3), and paraoxonase-1 and paraoxonase-3 (PON1, PON3), may be a feature of AD, even at the early stage. Moreover, the decrease in these enzymes with antioxidant/defensive action appears to be accompanied by a parallel increase of prooxidant and proinflammatory mediators, in particular myeloperoxidase (MPO) and serum amyloid A (SAA). This type of derangement of balance between two opposite forces makes HDL dysfunctional, i.e., unable to exert its "natural" vasculoprotective property. In this review, we summarized and critically analyzed the most significant findings linking HDL accessory proteins and AD. We also discuss the most convincing hypothesis explaining the mechanism by which an observed systemic occurrence may have repercussions in the brain.
Collapse
Affiliation(s)
- Francesca Zimetti
- Department of Food and Drug, University of Parma, Parma 43124, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neurosciences, University of Parma, Parma 43121, Italy
| | - Judit Marsillach
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Cinzia Marchi
- Department of Food and Drug, University of Parma, Parma 43124, Italy
| | - Alessandro Trentini
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara 44121, Italy
- Plants for Human Health Institute, Animal Sciences Department, NC Research Campus Kannapolis, NC State University, 28081 NC, USA
- Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea
| | - Carlo Cervellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
30
|
Di Costanzo A, Ronca A, D’Erasmo L, Manfredini M, Baratta F, Pastori D, Di Martino M, Ceci F, Angelico F, Del Ben M, Pavanello C, Turri M, Calabresi L, Favari E, Arca M. HDL-Mediated Cholesterol Efflux and Plasma Loading Capacities Are Altered in Subjects with Metabolically- but Not Genetically Driven Non-Alcoholic Fatty Liver Disease (NAFLD). Biomedicines 2020; 8:625. [PMID: 33352841 PMCID: PMC7766839 DOI: 10.3390/biomedicines8120625] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023] Open
Abstract
Background. Non-alcoholic fatty liver disease (NAFLD) increases the risk of atherosclerosis but this risk may differ between metabolically- vs. genetically-driven NAFLD. High-density lipoprotein (HDL)-mediated cholesterol efflux (CEC) and plasma loading capacity (CLC) are key factors in atherogenesis. Aims. To test whether CEC and CLC differ between metabolically- vs. genetically-determined NAFLD. Methods: CEC and CLC were measured in 19 patients with metabolic NAFLD and wild-type PNPLA3 genotype (Group M), 10 patients with genetic NAFLD carrying M148M PNPLA3 genotype (Group G), and 10 controls PNPLA3 wild-types and without NAFLD. CEC and CLC were measured ex vivo by isotopic and fluorimetric techniques using cellular models. Results: Compared with Group G, Group M showed reduced total CEC (-18.6%; p < 0.001) as well as that mediated by cholesterol transporters (-25.3% ABCA1; -16.3% ABCG1; -14.8% aqueous diffusion; all p < 0.04). No difference in CEC was found between Group G and controls. The presence of metabolic syndrome further impaired ABCG1-mediated CEC in Group M. Group M had higher plasma-induced CLC than Group G and controls (p < 0.001). Conclusions: Metabolically-, but not genetically-, driven NAFLD associates with dysfunctional HDL-meditated CEC and abnormal CLC. These data suggest that the mechanisms of anti-atherogenic protection in metabolic NAFLD are impaired.
Collapse
Affiliation(s)
- Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.D.); (M.A.)
| | - Annalisa Ronca
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (A.R.); (E.F.)
| | - Laura D’Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.D.); (M.A.)
| | - Matteo Manfredini
- Department of Chemistry, Life Science, and Environmental Sustainability, University of Parma, 43124 Parma, Italy;
| | - Francesco Baratta
- Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.B.); (D.P.); (M.D.B.)
| | - Daniele Pastori
- Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.B.); (D.P.); (M.D.B.)
| | - Michele Di Martino
- Department of Diagnostic of Radiological, Oncological and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Fabrizio Ceci
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesco Angelico
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Del Ben
- Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.B.); (D.P.); (M.D.B.)
| | - Chiara Pavanello
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (M.T.); (L.C.)
| | - Marta Turri
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (M.T.); (L.C.)
| | - Laura Calabresi
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (M.T.); (L.C.)
| | - Elda Favari
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (A.R.); (E.F.)
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.D.); (M.A.)
| |
Collapse
|
31
|
Nazir S, Jankowski V, Bender G, Zewinger S, Rye KA, van der Vorst EP. Interaction between high-density lipoproteins and inflammation: Function matters more than concentration! Adv Drug Deliv Rev 2020; 159:94-119. [PMID: 33080259 DOI: 10.1016/j.addr.2020.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 09/20/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
High-density lipoprotein (HDL) plays an important role in lipid metabolism and especially contributes to the reverse cholesterol transport pathway. Over recent years it has become clear that the effect of HDL on immune-modulation is not only dependent on HDL concentration but also and perhaps even more so on HDL function. This review will provide a concise general introduction to HDL followed by an overview of post-translational modifications of HDL and a detailed overview of the role of HDL in inflammatory diseases. The clinical potential of HDL and its main apolipoprotein constituent, apoA-I, is also addressed in this context. Finally, some conclusions and remarks that are important for future HDL-based research and further development of HDL-focused therapies are discussed.
Collapse
|
32
|
Favari E, Angelino D, Cipollari E, Adorni MP, Zimetti F, Bernini F, Ronda N, Pellegrini N. Functional pasta consumption in healthy volunteers modulates ABCG1-mediated cholesterol efflux capacity of HDL. Nutr Metab Cardiovasc Dis 2020; 30:1768-1776. [PMID: 32605885 DOI: 10.1016/j.numecd.2020.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/17/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUNDS AND AIMS Prevention of cardiovascular (CV) disease is considered a central issue in public health and great attention is payed to nutritional approaches, including consumption of functional foods to reduce CV risk in individuals without indications for anti-atherosclerotic drugs. Cholesterol efflux capacity (CEC) is an important anti-atherogenic property of HDL and a marker of CV risk. We evaluated the effect of a daily consumption of an innovative whole-wheat synbiotic pasta, compared to a control whole-wheat pasta, on serum ATP binding cassette G1 (ABCG1)-mediated CEC in healthy overweight or obese individuals. METHODS AND RESULTS Study participants (n = 41) were randomly allocated to either innovative or control pasta, consumed daily for twelve weeks. Serum CEC was measured before and after the dietary intervention, by a well-established radioisotopic technique on Chinese Hamster Ovary Cells transfected with human ABCG1. The innovative synbiotic pasta consumption was associated to a significantly higher post treatment/baseline ratio of ABCG1-mediated CEC values with respect to control pasta (mean ratio 1.05 ± 0.037 and 0.95 ± 0.042 respectively, p < 0.05). Analysis of the relationship between ABCG1-mediated CEC and glycemia, homocysteine, total folates and interleukin-6 showed specific changes in the correlations between HDL function and glycemia, oxidative and inflammatory markers only after synbiotic pasta consumption. CONCLUSION This is the first report on serum CEC improvement obtained by a new synbiotic functional pasta consumption, in absence of lipid profile modifications, in overweight/obese participants. This pilot study suggests that a simple dietary intervention can be a promising approach to CV preservation through improving of athero-protective HDL function.
Collapse
Affiliation(s)
- Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Donato Angelino
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | | | | | - Franco Bernini
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy.
| | | |
Collapse
|
33
|
Anti-atherogenic Modification of Serum Lipoprotein Function in Patients with Rheumatoid Arthritis after Tocilizumab Treatment, a Pilot Study. J Clin Med 2020; 9:jcm9072157. [PMID: 32650513 PMCID: PMC7408911 DOI: 10.3390/jcm9072157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Lipid metabolism derangement contributes to increased cardiovascular risk in Rheumatoid Arthritis (RA). It is still debated whether and how tocilizumab, an interleukin-6 receptor inhibitor used in active RA, impacts cardiovascular risk. We studied the effect of tocilizumab on the regulation of macrophage cholesterol homeostasis, measuring patient serum ability to respectively load (cholesterol loading capacity, CLC) and discharge (cholesterol efflux capacity, CEC) cells with cholesterol. Patients with RA (n = 8) were studied before and after 4 and 12 weeks of tocilizumab treatment. CLC was measured by a fluorimetric assay of intracellular cholesterol content in human macrophages and CEC was measured for the three main pathways, mediated by the transporters Scavenger Receptor class B-type I (SR-BI), ATP binding cassette-G1 (ABCG1) and -A1 (ABCA1) in specific cell models. After 12 weeks of tocilizumab treatment, serum LDL cholesterol levels were increased, while CLC was reduced. HDL cholesterol levels were unchanged, but CEC was significantly ameliorated for the SR-BI and ABCG1 pathways with respect to baseline. Tocilizumab reduces LDL pro-atherogenic potential despite increasing their serum levels and increases HDL protective activity in RA. The data of our pilot study suggest that tocilizumab regulates lipoprotein function in selected patient populations and lay the groundwork for future larger studies.
Collapse
|
34
|
Hafiane A, Favari E, Daskalopoulou SS, Vuilleumier N, Frias MA. High-density lipoprotein cholesterol efflux capacity and cardiovascular risk in autoimmune and non-autoimmune diseases. Metabolism 2020; 104:154141. [PMID: 31923386 DOI: 10.1016/j.metabol.2020.154141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/22/2022]
Abstract
Functional assessment of cholesterol efflux capacity (CEC) to high-density lipoprotein (HDL) is an emerging tool for evaluating morbidity and mortality associated with cardiovascular disease (CVD). By promoting macrophage reverse cholesterol transport (RCT), HDL-mediated CEC is believed to play an important role in atherosclerotic lesion progression in the vessel wall. Furthermore, recent evidence indicates that the typical inverse associations between various forms of CEC and CV events may be strongly modulated by environmental systemic factors and traditional CV risk factors, in addition to autoimmune diseases. These factors influence the complex and dynamic composition of HDL particles, which in turn positively or negatively affect HDL-CEC. Herein, we review recent findings connecting HDL-CEC to traditional CV risk factors and cardiometabolic conditions (non-autoimmune diseases) as well as autoimmune diseases, with a specific focus on how these factors may influence the associations between HDL-CEC and CVD risk.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01. 3370H, Montréal, Qc H4A 3J1, Canada.
| | - Elda Favari
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy.
| | - Stella S Daskalopoulou
- Department of Medicine, Division of Internal Medicine, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, EM1.2230, Montreal, Quebec H4A 3J1, Canada.
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; Division of Laboratory Medicine, Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| | - Miguel A Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; Division of Laboratory Medicine, Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| |
Collapse
|
35
|
Li J, Zhou C, Xu H, Brook RD, Liu S, Yi T, Wang Y, Feng B, Zhao M, Wang X, Zhao Q, Chen J, Song X, Wang T, Liu S, Zhang Y, Wu R, Gao J, Pan B, Pennathur S, Rajagopalan S, Huo Y, Zheng L, Huang W. Ambient Air Pollution Is Associated With HDL (High-Density Lipoprotein) Dysfunction in Healthy Adults. Arterioscler Thromb Vasc Biol 2020; 39:513-522. [PMID: 30700134 DOI: 10.1161/atvbaha.118.311749] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective- We aimed to assess whether exposure to higher levels of ambient air pollution impairs HDL (high-density lipoprotein) function and to elucidate the underlying biological mechanisms potentially involved. Approach and Results- In the Beijing AIRCHD study (Air Pollution and Cardiovascular Dysfunction in Healthy Adults), 73 healthy adults (23.3±5.4 years) were followed-up with 4 repeated study visits in 2014 to 2016. During each visit, ambient air pollution concentrations, HDL function metrics, and parameters of inflammation and oxidative stress were measured. Average daily concentrations of ambient particulate matter in diameter <2.5 μm were 62.9 µg/m3 (8.1-331.0 µg/m3). We observed significant decreases in HDL cholesterol efflux capacity of 2.3% (95% CI, -4.3 to -0.3) to 5.0% (95% CI, -7.6 to -2.4) associated with interquartile range increases in moving average concentrations of particulate matter in diameter <2.5 μm and traffic-related air pollutants (black carbon, nitrogen dioxide, and carbon monoxide) during the 1 to 7 days before each participant's clinic visit. Higher ambient air pollutant levels were also associated with significant reductions in circulating HDL cholesterol and apoA-I (apolipoprotein A-I), as well as elevations in HDL oxidation index, oxidized LDL (low-density lipoprotein), malondialdehyde, and high-sensitivity C-reactive protein. Conclusions- Higher ambient air pollution concentrations were associated with impairments in HDL functionality, potentially because of systemic inflammation and oxidative stress. These novel findings further our understanding of the mechanisms whereby air pollutants promote cardiometabolic disorders.
Collapse
Affiliation(s)
- Jianping Li
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Changping Zhou
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Hongbing Xu
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Robert D Brook
- Division of Cardiovascular Medicine (R.D.B.), University of Michigan, Ann Arbor
| | - Shengcong Liu
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Tieci Yi
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.)
| | - Yang Wang
- Department of Prevention and Health Care, Hospital of Health Science Center (Y.W.), Peking University, Beijing
| | - Baihuan Feng
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.)
| | - Mingming Zhao
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing
| | - Xu Wang
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing
| | - Qian Zhao
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.)
| | - Jie Chen
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Institute for Risk Assessment Sciences (J.C.), University Medical Centre Utrecht, University of Utrecht, the Netherlands.,Julius Centre for Health Sciences and Primary Care (J.C.), University Medical Centre Utrecht, University of Utrecht, the Netherlands
| | - Xiaoming Song
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Tong Wang
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Shuo Liu
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.)
| | - Yi Zhang
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Rongshan Wu
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Jianing Gao
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Bing Pan
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | | | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Case Western Reserve Medical School, Cleveland OH (S.R.), Peking University, Beijing
| | - Yong Huo
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Lemin Zheng
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Wei Huang
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| |
Collapse
|
36
|
Bonacina F, Pirillo A, Catapano AL, Norata GD. Cholesterol membrane content has a ubiquitous evolutionary function in immune cell activation: the role of HDL. Curr Opin Lipidol 2019; 30:462-469. [PMID: 31577612 DOI: 10.1097/mol.0000000000000642] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Cellular cholesterol content influences the structure and function of lipid rafts, plasma membrane microdomains essential for cell signaling and activation. HDL modulate cellular cholesterol efflux, thus limiting cholesterol accumulation and controlling immune cell activation. Aim of this review is to discuss the link between HDL and cellular cholesterol metabolism in immune cells and the therapeutic potential of targeting cholesterol removal from cell membranes. RECENT FINDINGS The inverse relationship between HDL-cholesterol (HDL-C) levels and the risk of cardiovascular disease has been recently challenged by observations linking elevated levels of HDL-C with increased risk of all-cause mortality, infections and autoimmune diseases, paralleled by the failure of clinical trials with HDL-C-raising therapies. These findings suggest that improving HDL function might be more important than merely raising HDL-C levels. New approaches aimed at increasing the ability of HDL to remove cellular cholesterol have been assessed for their effect on immune cells, and the results have suggested that this could be a new effective approach. SUMMARY Cholesterol removal from plasma membrane by different means affects the activity of immune cells, suggesting that approaches aimed at increasing the ability of HDL to mobilize cholesterol from cells would represent the next step in HDL biology.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital
- IRCCS MultiMedica, Milan, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan
- IRCCS MultiMedica, Milan, Italy
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan
- Center for the Study of Atherosclerosis, E. Bassini Hospital
| |
Collapse
|
37
|
Adorni MP, Zimetti F, Cangiano B, Vezzoli V, Bernini F, Caruso D, Corsini A, Sirtori CR, Cariboni A, Bonomi M, Ruscica M. High-Density Lipoprotein Function Is Reduced in Patients Affected by Genetic or Idiopathic Hypogonadism. J Clin Endocrinol Metab 2019; 104:3097-3107. [PMID: 30835274 DOI: 10.1210/jc.2018-02027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/26/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Low testosterone levels are associated with an increased incidence of cardiovascular (CV) events, but the underlying biochemical mechanisms are not fully understood. The clinical condition of hypogonadism offers a unique model to unravel the possible role of lipoprotein-associated abnormalities in CV risk. In particular, the assessment of the functional capacities of high-density lipoproteins (HDLs) may provide insights besides traditional risk factors. DESIGN To determine whether reduced testosterone levels correlate with lipoprotein function, HDL cholesterol (HDL-C) efflux capacity (CEC) and serum cholesterol loading capacity (CLC). PARTICIPANTS Genetic and idiopathic hypogonadal patients (n = 20) and control subjects (n = 17). RESULTS Primary and secondary hypogonadal patients presented with lower HDL ATP-binding cassette transporter A1 (ABCA1)-, ATP-binding cassette transporter G1 (ABCG1)-, and aqueous diffusion-mediated CEC (-19.6%, -40.9%, and -12.9%, respectively), with a 16.2% decrement of total CEC. In the whole series, positive correlations between testosterone levels and both total HDL CEC (r2 = 0.359, P = 0.0001) and ABCG1 HDL CEC (r2 = 0.367, P = 0.0001) were observed. Conversely, serum CLC was markedly raised (+43%) in hypogonadals, increased, to a higher extent, in primary vs secondary hypogonadism (18.45 ± 2.78 vs 15.15 ± 2.10 µg cholesterol/mg protein) and inversely correlated with testosterone levels (r2 = 0.270, P = 0.001). HDL-C concentrations did not correlate with either testosterone levels, HDL CEC (total, ABCG1, and ABCA1) or serum CLC. CONCLUSIONS In hypogonadal patients, proatherogenic lipoprotein-associated changes are associated with lower cholesterol efflux and increased influx, thus offering an explanation for a potentially increased CV risk.
Collapse
Affiliation(s)
| | | | - Biagio Cangiano
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Laboratory of Endocrine and Metabolic Research and Division of Endocrine and Metabolic Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Auxologico Italiano, Milan, Italy
| | - Valeria Vezzoli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Laboratory of Endocrine and Metabolic Research and Division of Endocrine and Metabolic Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Auxologico Italiano, Milan, Italy
| | - Franco Bernini
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Multimedica Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy
| | - Cesare R Sirtori
- Centro Dislipidemie, Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marco Bonomi
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Laboratory of Endocrine and Metabolic Research and Division of Endocrine and Metabolic Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Auxologico Italiano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
38
|
Pirillo A, Catapano AL, Norata GD. Biological Consequences of Dysfunctional HDL. Curr Med Chem 2019; 26:1644-1664. [PMID: 29848265 DOI: 10.2174/0929867325666180530110543] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/25/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022]
Abstract
Epidemiological studies have suggested an inverse correlation between high-density lipoprotein (HDL) cholesterol levels and the risk of cardiovascular disease. HDLs promote reverse cholesterol transport (RCT) and possess several putative atheroprotective functions, associated to the anti-inflammatory, anti-thrombotic and anti-oxidant properties as well as to the ability to support endothelial physiology. The assumption that increasing HDL-C levels would be beneficial on cardiovascular disease (CVD), however, has been questioned as, in most clinical trials, HDL-C-raising therapies did not result in improved cardiovascular outcomes. These findings, together with the observations from Mendelian randomization studies showing that polymorphisms mainly or solely associated with increased HDL-C levels did not decrease the risk of myocardial infarction, shift the focus from HDL-C levels toward HDL functional properties. Indeed, HDL from atherosclerotic patients not only exhibit impaired atheroprotective functions but also acquire pro-atherogenic properties and are referred to as "dysfunctional" HDL; this occurs even in the presence of normal or elevated HDL-C levels. Pharmacological approaches aimed at restoring HDL functions may therefore impact more significantly on CVD outcome than drugs used so far to increase HDL-C levels. The aim of this review is to discuss the pathological conditions leading to the formation of dysfunctional HDL and their role in atherosclerosis and beyond.
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,IRCCS Multimedica, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Danilo Norata
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia
| |
Collapse
|
39
|
Marchi C, Adorni MP, Caffarra P, Ronda N, Spallazzi M, Barocco F, Galimberti D, Bernini F, Zimetti F. ABCA1- and ABCG1-mediated cholesterol efflux capacity of cerebrospinal fluid is impaired in Alzheimer's disease. J Lipid Res 2019; 60:1449-1456. [PMID: 31167810 DOI: 10.1194/jlr.p091033] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/25/2019] [Indexed: 01/18/2023] Open
Abstract
HDL-like particles in human cerebrospinal fluid (CSF) promote the efflux of cholesterol from astrocytes toward the neurons that rely on this supply for their functions. We evaluated whether cell cholesterol efflux capacity of CSF (CSF-CEC) is impaired in Alzheimer's disease (AD) by analyzing AD (n = 37) patients, non-AD dementia (non-AD DEM; n = 16) patients, and control subjects (n = 39). As expected, AD patients showed reduced CSF Aβ 1-42, increased total and phosphorylated tau, and a higher frequency of the apoε4 genotype. ABCA1- and ABCG1-mediated CSF-CEC was markedly reduced in AD (-73% and -33%, respectively) but not in non-AD DEM patients, in which a reduced passive diffusion CEC (-40%) was observed. Non-AD DEM patients displayed lower CSF apoE concentrations (-24%) compared with controls, while apoA-I levels were similar among groups. No differences in CSF-CEC were found by stratifying subjects for apoε4 status. ABCG1 CSF-CEC positively correlated with Aβ 1-42 (r = 0.305, P = 0.025), while ABCA1 CSF-CEC inversely correlated with total and phosphorylated tau (r = -0.348, P = 0.018 and r = -0.294, P = 0.048, respectively). The CSF-CEC impairment and the correlation with the neurobiochemical markers suggest a pathophysiological link between CSF HDL-like particle dysfunction and neurodegeneration in AD.
Collapse
Affiliation(s)
- Cinzia Marchi
- Department of Food and Drug University of Parma, Parma, Italy
| | | | - Paolo Caffarra
- Department of Medicine and Surgery, Section of Neurology University of Parma, Parma, Italy.,Alzheimer Center Briolini Hospital, Gazzaniga, Bergamo, Italy
| | - Nicoletta Ronda
- Department of Food and Drug University of Parma, Parma, Italy
| | - Marco Spallazzi
- Department of Medicine and Surgery, Section of Neurology University of Parma, Parma, Italy
| | - Federica Barocco
- Department of Medicine and Surgery, Section of Neurology University of Parma, Parma, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, University of Milano, Milano, Italy.,Neurodegenerative Diseases Unit Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Franco Bernini
- Department of Food and Drug University of Parma, Parma, Italy
| | | |
Collapse
|
40
|
A proteomic approach to identify novel disease biomarkers in LCAT deficiency. J Proteomics 2018; 198:113-118. [PMID: 30529744 DOI: 10.1016/j.jprot.2018.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 12/22/2022]
Abstract
Genetic LCAT deficiency is a rare recessive autosomal disease due to loss-of-function mutations in the gene coding for the enzyme lecithin:cholesterol acyltransferase (LCAT). Homozygous carriers are characterized by corneal opacity, haemolytic anaemia and renal disease, which represent the first cause of morbidity and mortality in these subjects. Diagnostic and prognostic markers capable of early detecting declining kidney function in these subjects are not available, and the specific serum or urine proteomic signature of LCAT deficient carriers has never been assessed. Taking advantage of a proteomic approach, we performed 2-DE analysis of carriers' plasma and identified proteins present at different concentration in samples from homozygous carriers. Our data confirm the well-known alterations in the concentration of circulating apolipoproteins, with a statistically significant decrease of both apoA-I and apoA-II and a statistically significant increase of apoC-III. Furthermore, we observed increased level of alpha-1-antitrypsin, zinc-alpha-2-glycoprotein and retinol-binding protein 4, and reduced level of clusterin and haptoglobin. Interestingly, only beta but not alpha subunit of haptoglobin is significant reduced in homozygous subjects. Despite the limited sample size, our findings set the basis for assessing the identified protein in a larger population and for correlating their levels with clinical markers of renal function and anaemia. SIGNIFICANCE: This investigation defines the effects of LCAT deficiency on the level of the major plasma proteins in homozygous and heterozygous carriers. Increase for some proteins, with different function, together with a drop for haptoglobin, and specifically for haptoglobin beta chains, are reported for the first time as part of a coherent signature. We are glad to have the opportunity to report our findings on this subject, which is one of the main interests for our research group, when Journal of Proteomics celebrates its 10th anniversary. With its various sections devoted to different areas of research, this journal is a privileged forum for publishing proteomic investigations without restrictions either in sample type or in technical approach. It is as well a privileged forum for reviewing literature data on various topics related to proteomics investigation, as colleagues in our research group have done over the years; by the way, a good share of the reviewed papers were as well reports published in Journal of Proteomics itself. The journal also offers opportunities for focused surveys through thematic issues devoted to a variety of subjects, timely selected for their current relevance in research; it was an honour for colleagues in our group to recently act as editors of one of those. Out of this diverse experience, we express our appreciation for the endeavour of Journal of Proteomics in its first 10 years of life - and wish identical and possibly greater success for the time to come.
Collapse
|
41
|
Schulte DM, Paulsen K, Türk K, Brandt B, Freitag-Wolf S, Hagen I, Zeuner R, Schröder JO, Lieb W, Franke A, Nikolaus S, Mrowietz U, Gerdes S, Schreiber S, Laudes M. Small dense LDL cholesterol in human subjects with different chronic inflammatory diseases. Nutr Metab Cardiovasc Dis 2018; 28:1100-1105. [PMID: 30143407 DOI: 10.1016/j.numecd.2018.06.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/06/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND AND AIMS Chronic inflammatory diseases (CID) are associated with a profound increase in cardiovascular (CV) risk resulting in reduced life expectancy. However, LDL-cholesterol is reported to be low in CID patients which is referred to as the "LDL paradoxon". The aim of the present study was to investigate whether LDL-particles in CID exhibit an increased content of the highly atherogenic small-dense LDL subfraction (sdLDL). METHODS AND RESULTS In this prospective, single center, observational study we enrolled 141 patients with CID (RA n = 59, inflammatory bowel disease (IBD) n = 35, ankylosing spondylitis (SpA) n = 25, Psoriasis n = 22) in 2011 through 2013 to evaluate sdLDL levels before as well as 6 and 26 weeks after initiation of different anti-cytokine therapies (anti-TNFα, anti-IL-6R antibodies). sdLDL levels were compared to 141 healthy individuals in a case control design. Compared to healthy controls, all CID patients displayed a significantly higher sdLDL content within the LDL cholesterol fraction: RA 35.0 ± 9.2% (p < 0.001), SpA 42.5 ± 10.5% (p < 0.001), IBD 37.5 ± 7.1% (p < 0.001), Psoriasis 33.6 ± 4.6% (p < 0.01). Furthermore, the sdLDL/LDL ratio was significantly higher in male compared to female RA subjects (p < 0.05). Neither anti-TNFα nor anti-IL6R medication altered sdLDL levels despite a significant improvement of disease activity. CONCLUSION In several different chronic inflammatory disease entities, LDL-cholesterol is shifted toward a pro-atherogenic phenotype due to an increased sdLDL content which might in part explain the LDL paradoxon. Since premature CV disease is a major burden of affected patients, specifically targeting lipid metabolism should be considered routinely in clinical patient care. CLINICAL TRIALS Registration at German Clinical Trial Register (DRKS): DRKS00005285.
Collapse
Affiliation(s)
- D M Schulte
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - K Paulsen
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - K Türk
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - B Brandt
- Institute of Clinical Chemistry, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Freitag-Wolf
- Institute of Medical Informatics and Statistics, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - I Hagen
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - R Zeuner
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - J O Schröder
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - W Lieb
- Institute of Epidemiology, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - A Franke
- Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Institute of Clinical Molecular Biology, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Nikolaus
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - U Mrowietz
- Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Psoriasis-Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Gerdes
- Psoriasis-Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Schreiber
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - M Laudes
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany.
| |
Collapse
|
42
|
Contreras-Duarte S, Chen P, Andía M, Uribe S, Irarrázaval P, Kopp S, Kern S, Marsche G, Busso D, Wadsack C, Rigotti A. Attenuation of atherogenic apo B-48-dependent hyperlipidemia and high density lipoprotein remodeling induced by vitamin C and E combination and their beneficial effect on lethal ischemic heart disease in mice. Biol Res 2018; 51:34. [PMID: 30219096 PMCID: PMC6138920 DOI: 10.1186/s40659-018-0183-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/06/2018] [Indexed: 01/01/2023] Open
Abstract
Background and aims Atherosclerotic cardiovascular disease is highly prevalent and its underlying pathogenesis involves dyslipidemia including pro-atherogenic high density lipoprotein (HDL) remodeling. Vitamins C and E have been proposed as atheroprotective agents for cardiovascular disease management. However, their effects and benefits on high density lipoprotein function and remodeling are unknown. In this study, we evaluated the role of vitamin C and E on non HDL lipoproteins as well as HDL function and remodeling, along with their effects on inflammation/oxidation biomarkers and atherosclerosis in atherogenic diet-fed SR-B1 KO/ApoER61h/h mice. Methods and results Mice were pre-treated for 5 weeks before and during atherogenic diet feeding with vitamin C and E added to water and diet, respectively. Compared to a control group, combined vitamin C and E administration reduced serum total cholesterol and triglyceride levels by decreasing apo B-48-containing lipoproteins, remodeled HDL particles by reducing phospholipid as well as increasing PON1 and apo D content, and diminished PLTP activity and levels. Vitamin supplementation improved HDL antioxidant function and lowered serum TNF-α levels. Vitamin C and E combination attenuated atherogenesis and increased lifespan in atherogenic diet-fed SR-B1 KO/ApoER61h/h mice. Conclusions Vitamin C and E administration showed significant lipid metabolism regulating effects, including HDL remodeling and decreased levels of apoB-containing lipoproteins, in mice. In addition, this vitamin supplementation generated a cardioprotective effect in a murine model of severe and lethal atherosclerotic ischemic heart disease.
Collapse
Affiliation(s)
- S Contreras-Duarte
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay #362 - 4º, Piso, 8330024, Santiago, Chile
| | - P Chen
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay #362 - 4º, Piso, 8330024, Santiago, Chile
| | - M Andía
- Department of Radiology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Biomedical Imaging Center, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Electrical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Uribe
- Department of Radiology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Biomedical Imaging Center, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Electrical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - P Irarrázaval
- Biomedical Imaging Center, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Electrical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Kopp
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - S Kern
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - G Marsche
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - D Busso
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay #362 - 4º, Piso, 8330024, Santiago, Chile
| | - C Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - A Rigotti
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Diagonal Paraguay #362 - 4º, Piso, 8330024, Santiago, Chile. .,Center of Molecular Nutrition and Chronic Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
43
|
Guirgis FW, Dodani S, Leeuwenburgh C, Moldawer L, Bowman J, Kalynych C, Grijalva V, Reddy ST, Jones AE, Moore FA. HDL inflammatory index correlates with and predicts severity of organ failure in patients with sepsis and septic shock. PLoS One 2018; 13:e0203813. [PMID: 30216360 PMCID: PMC6138388 DOI: 10.1371/journal.pone.0203813] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
Objective High density lipoprotein (HDL) is important for defense against sepsis but becomes dysfunctional (Dys-HDL) during inflammation. We hypothesize that Dys-HDL correlates with organ dysfunction (sequential organ failure assessment (SOFA) score) early sepsis. Methods A prospective cohort study of adult ED sepsis patients enrolled within 24 hours. Results Eighty eight patients were analyzed. Dys-HDL (expressed as HDL inflammatory index (HII)) correlated with SOFA at enrollment (r = 0.23, p = 0.024) and at 48 hours (r = 0.24, p = 0.026) but HII change over the first 48 hours did not correlate with change in SOFA (r = 0.06, p = 0.56). Enrollment HII was significantly different in patients with most severe organ failure (2.31, IQR 1.33–5.2) compared to less severe organ failure (1.81, IQR 1.23–2.64, p = 0.043). Change in HII over 48 hours was significantly different for in-hospital non-survivors (-0.45, IQR-2.6, -0.14 p = 0.015) and for 28-day non-survivors (-1.12, IQR -1.52, 0.12, p = 0.044). In a multivariable linear regression equation (R2 = 0.13), for each unit HII increase, 48-hour SOFA increased by 0.72 (p = 0.009). Conclusion HII correlated with SOFA and predicted 48-hour SOFA score in early sepsis. Future studies are needed to delineate potential mechanisms. Trial registration NCT02370186. Registered February 24, 2015.
Collapse
Affiliation(s)
- Faheem W. Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, United States of America
- * E-mail:
| | - Sunita Dodani
- Department of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States of America
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatrics, University of Florida, College of Medicine, Gainesville, FL, United States of America
| | - Lyle Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States of America
| | - Jennifer Bowman
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, United States of America
| | - Colleen Kalynych
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, United States of America
| | - Victor Grijalva
- Department of Medicine, Molecular & Medical Pharmacology, UCLA School of Medicine, Los Angeles, CA, United States of America
| | - Srinivasa T. Reddy
- Department of Medicine, Molecular & Medical Pharmacology, UCLA School of Medicine, Los Angeles, CA, United States of America
| | - Alan E. Jones
- Department of Emergency Medicine, University of Mississippi College of Medicine, Jackson, MS, United States of America
| | - Frederick A. Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States of America
| |
Collapse
|
44
|
Kaseda R, Tsuchida Y, Gamboa JL, Zhong J, Zhang L, Yang H, Dikalova A, Bian A, Davies S, Fogo AF, Linton MF, Brown NJ, Ikizler TA, Kon V. Angiotensin receptor blocker vs ACE inhibitor effects on HDL functionality in patients on maintenance hemodialysis. Nutr Metab Cardiovasc Dis 2018; 28:582-591. [PMID: 29691148 PMCID: PMC5959764 DOI: 10.1016/j.numecd.2018.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Angiotensin receptor blockers (ARB) and angiotensin converting enzyme inhibitors (ACEI) reduce cardiovascular events in the general population. Maintenance hemodialysis (MHD) patients are at high cardiovascular risk but few studies have directly addressed the comparative efficacy of these drugs. MHD disrupts the normally atheroprotective actions of high density lipoprotein (HDL), therefore, we compared ACEI or ARB treatment on HDL functions in MHD. METHODS AND RESULTS HDL was isolated at the starting point (pre) and 3-6 months later (post) in 30 MHD randomly assigned to placebo, ramipril or valsartan. Outcomes included cholesterol efflux, inflammatory cytokine response, effects on Toll-like receptors (TLR), superoxide production, methylarginine and serum amyloid A (SAA) levels. HDL from ARB- or ACEI-treated subjects was more effective in maintaining efflux than HDL of placebo. HDL from ARB- or ACEI-treated subjects but not placebo lessened cellular superoxide production. In contrast, neither ARB nor ACEI improved HDL anti-inflammatory effect. Indeed, HDL of ACEI-treated subjects potentiated the cytokine responses in association with activation of TLR but did not alter the HDL content of methylarginines or SAA. CONCLUSION Both ACEI and ARB stabilized HDL cholesterol acceptor function and sustained cellular anti-oxidative effects but not anti-inflammatory effects, and ACEI-treatment instead amplified the HDL inflammatory response. The findings reveal possible utility of antagonizing angiotensin actions in MDH and suggest a possible mechanism for superiority of ARB vs ACEI in the setting of advanced kidney disease.
Collapse
Affiliation(s)
- R Kaseda
- Pediatric Nephrology, VUMC, Nashville, TN, USA
| | - Y Tsuchida
- Pediatric Nephrology, VUMC, Nashville, TN, USA
| | | | - J Zhong
- Pediatric Nephrology, VUMC, Nashville, TN, USA; Pathology, Microbiology and Immunology, Nashville, TN, USA
| | - L Zhang
- Pharmacology, Nashville, TN, USA
| | - H Yang
- Pediatric Nephrology, VUMC, Nashville, TN, USA; Pathology, Microbiology and Immunology, Nashville, TN, USA
| | | | - A Bian
- Biostatistics, Vanderbilt Medical Center, Nashville, TN, USA
| | - S Davies
- Pharmacology, Nashville, TN, USA
| | - A F Fogo
- Pediatric Nephrology, VUMC, Nashville, TN, USA; Medicine, Nashville, TN, USA; Pathology, Microbiology and Immunology, Nashville, TN, USA
| | | | | | | | - V Kon
- Pediatric Nephrology, VUMC, Nashville, TN, USA.
| |
Collapse
|