1
|
Bae G, Yang Z, Bucci D, Wegner C, Schäfer H, Singh Y, Lonati C, Trautwein C. Longitudinal lipoprotein and inflammatory mediators analysis uncover persisting inflammation and hyperlipidemia following SARS-CoV-2 infection in long COVID-19. Metabolomics 2025; 21:65. [PMID: 40335840 PMCID: PMC12058914 DOI: 10.1007/s11306-025-02262-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/18/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION Individuals suffering from acute COVID-19 (AC) often develop long COVID-19 (LC) syndrome that is associated with aberrant levels of lipoproteins and inflammatory mediators. Yet, these dysregulations are heterogenous due to the uncertain prevalence and require a more extensive characterization. OBJECTIVES This study aimed to investigate LC-associated dysregulations in inflammatory mediators and lipids by longitudinal Nuclear Magnetic Resonance (NMR) lipoprotein analysis and cytokine profiling in human blood. METHODS We quantitatively profiled lipoproteins and inflammatory parameters in LC patients at 5 (n = 95), 9 (n = 73), 12 (n = 95), 16 (n = 78), and 20 (n = 85) months post AC by in vitro diagnostics research (IVDr)-based NMR spectroscopy. Simultaneously, we assessed inflammatory meditators with a 13-plex cytokine panel by flow cytometry. We then compared the lipoprotein profiles with historical data from AC (N = 307) and healthy cohorts collected before the COVID-19 pandemic (N = 305), whereas the cytokine profiles were correlated with that of the AC cohort. RESULTS We identified 31 main and 80 significantly altered subclass lipoproteins, respectively. LC was associated with higher serum levels of very low-density, intermediate-density, low-density, high-density lipoproteins, along with triglycerides, cholesterols, and apolipoprotein a-I & a-II lipoproteins compared to the healthy cohort. We also observed significantly lower concentrations of NMR-based inflammatory parameters in LC than in AC cohort, whilst proinflammatory mediators IFN-α2, IFN-γ, TNF-α, CXCL8/IL-8, IL-12p70, IL-17 A, and IL-23 displayed significantly higher concentrations in LC compared with the AC cohort. Conversely, CCL2/MCP-1, IL-6, and IL-18 were significantly higher in the AC cohort than in LC. CONCLUSION Our findings demonstrate a persistent hyperlipidemic phenotype in LC alongside signs of chronic inflammation and lipoprotein metabolism that vary in states of acute and chronic inflammation.
Collapse
Affiliation(s)
- Gyuntae Bae
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Zhiqi Yang
- Research Institute of Women's Health, University of Tübingen, Tübingen, Germany
| | - Daniele Bucci
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Claire Wegner
- Bruker BioSpin GmbH & Co. KG, Biopharma & Applied Division, Ettlingen, Germany
| | - Hartmut Schäfer
- Bruker BioSpin GmbH & Co. KG, Biopharma & Applied Division, Ettlingen, Germany
| | - Yogesh Singh
- Research Institute of Women's Health, University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Next Generation Sequencing (NGS) Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany.
- M3 Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany.
- Core Facility Metabolomics, Faculty of Medicine, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
2
|
Liu K, Cooper ME, Chai Z, Liu F. High-Density Lipoprotein in Patients with Diabetic Kidney Disease: Friend or Foe? Int J Mol Sci 2025; 26:1683. [PMID: 40004147 PMCID: PMC11855193 DOI: 10.3390/ijms26041683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
High-density lipoprotein (HDL) exhibits multiple metabolic protective functions, such as facilitating cellular cholesterol efflux, antioxidant, anti-inflammatory, anti-apoptotic and anti-thrombotic properties, showing antidiabetic and renoprotective potential. Diabetic kidney disease (DKD) is considered to be associated with high-density lipoprotein cholesterol (HDL-C). The hyperglycemic environment, non-enzymatic glycosylation, carbamylation, oxidative stress and systemic inflammation can cause changes in the quantity and quality of HDL, resulting in reduced HDL levels and abnormal function. Dysfunctional HDL can also have a negative impact on pancreatic β cells and kidney cells, leading to the progression of DKD. Based on these findings, new HDL-related DKD risk predictors have gradually been proposed. Interventions aiming to improve HDL levels and function, such as infusion of recombinant HDL (rHDL) or lipid-poor apolipoprotein A-I (apoA-I), can significantly improve glycemic control and also show renal protective effects. However, recent studies have revealed a U-shaped relationship between HDL-C levels and DKD, and the loss of protective properties of high levels of HDL may be related to changes in composition and the deposition of dysfunctional particles that exacerbate damage. Further research is needed to fully elucidate the complex role of HDL in DKD. Given the important role of HDL in metabolic health, developing HDL-based therapies that augment HDL function, rather than simply increasing its level, is a critical step in managing the development and progression of DKD.
Collapse
Affiliation(s)
- Ke Liu
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China;
- Laboratory of Diabetic Kidney Disease, Kidney Research Institute, Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mark E. Cooper
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia;
| | - Zhonglin Chai
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia;
| | - Fang Liu
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China;
- Laboratory of Diabetic Kidney Disease, Kidney Research Institute, Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Abidor E, Achkar M, Al Saidi I, Lather T, Jdaidani J, Agarwal A, El-Sayegh S. Comprehensive Review of Lipid Management in Chronic Kidney Disease and Hemodialysis Patients: Conventional Approaches, and Challenges for Cardiovascular Risk Reduction. J Clin Med 2025; 14:643. [PMID: 39860649 PMCID: PMC11765848 DOI: 10.3390/jcm14020643] [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: 12/09/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Lipid disorders are very prevalent in patients with chronic kidney disease (CKD) and end-stage kidney disease (ESKD), leading to heightened cardiovascular risk. This review examines the effectiveness of lipid-lowering agents in these populations and explores gaps in the current research. The goal of this review is to assess the efficacy of lipid-lowering therapies in CKD and ESRD patients and identify future research needs. It aims to provide a clearer understanding of how these treatments impact cardiovascular risk in high-risk populations. Methods: We conducted a literature search in Embase, PubMed, Cochrane, and Google Scholar databases using keywords including but not limited to: chronic kidney diseases, dialysis, hemodialysis, dyslipidemia, statins, ezetimibe, and lipid-lowering drugs. Findings from included studies were synthetized to provide an overview of the current management of dyslipidemia in ESRD and HD. Results: Statins show mixed results in CKD and ESRD, with limited benefits in reducing cardiovascular events in dialysis patients. Agents like PCSK9 inhibitors show promising results but require further research, while non-statin therapies like fibrates and omega-3 fatty acids have limited evidence for use in this population. Conclusions: The review underscores the need for further research into lipid-lowering agents in CKD and ESRD patients, highlighting the need for tailored lipid management strategies in vulnerable patients with unique risk factors. More studies are needed to refine treatment strategies and assess the role of exercise and accurate risk calculators in managing cardiovascular outcomes.
Collapse
Affiliation(s)
- Erica Abidor
- Department of Medicine, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA; (E.A.); (M.A.); (I.A.S.); (T.L.); (J.J.); (A.A.)
| | - Michel Achkar
- Department of Medicine, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA; (E.A.); (M.A.); (I.A.S.); (T.L.); (J.J.); (A.A.)
| | - Ibrahim Al Saidi
- Department of Medicine, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA; (E.A.); (M.A.); (I.A.S.); (T.L.); (J.J.); (A.A.)
| | - Tanvi Lather
- Department of Medicine, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA; (E.A.); (M.A.); (I.A.S.); (T.L.); (J.J.); (A.A.)
| | - Jennifer Jdaidani
- Department of Medicine, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA; (E.A.); (M.A.); (I.A.S.); (T.L.); (J.J.); (A.A.)
| | - Alaukika Agarwal
- Department of Medicine, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA; (E.A.); (M.A.); (I.A.S.); (T.L.); (J.J.); (A.A.)
| | - Suzanne El-Sayegh
- Department of Medicine, Division of Nephrology, Northwell Health, Staten Island University Hospital, Staten Island, NY 10305, USA
| |
Collapse
|
4
|
Dunne S, Gibney ER, McGillicuddy FC, Feeney EL. The effects of saturated fat intake from dairy on CVD markers: the role of food matrices. Proc Nutr Soc 2024; 83:236-244. [PMID: 38316603 DOI: 10.1017/s0029665124000132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
CVD is the leading cause of death worldwide, and is commonly associated with modifiable risk factors. Most studies to date examining link between food intake and risk of CVD, have focused on modulation of plasma cholesterol concentrations (total cholesterol (TC), LDL-C). However, recent studies suggest LDL particle size is a more sensitive risk marker for CVD with smaller, dense LDL particles reported as more atherogenic than larger, more buoyant LDL. Although dietary guidelines recommend SFA intake of < 10 % of total energy, this does not consider food source, with recent evidence suggesting differing, sometimes beneficial, lipid responses following consumption of SFA from dairy compared to other food sources. This may be from differences in the physical food matrices, the nutrient content of the foods, and/or how these components interact with each other, described as a 'dairy matrix effect'. Dietary fat not only raises LDL-C, but also HDL cholesterol (HDL-C), associated with reduced CVD risk. HDL particles are complex emulsions of lipids, proteins and microRNAs that exhibit atheroprotective properties. In addition, HDL particles exhibit a very heterogeneous proteomic composition, dependent on a person's disease state - with a more pro-inflammatory proteome evident in patients with established CVD. This review will discuss the evidence to date on the importance of the food matrix in modulating response to dietary SFA and impact on CVD risk factors. A focus on potential biomarker properties of lipoprotein particles beyond cholesterol and current use of such biomarkers in human nutrition research will be considered.
Collapse
Affiliation(s)
- Simone Dunne
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- UCD Institute for Food and Health, University College Dublin, Dublin 4, Republic of Ireland
| | - Eileen R Gibney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- UCD Institute for Food and Health, University College Dublin, Dublin 4, Republic of Ireland
| | - Fiona C McGillicuddy
- UCD Diabetes Complications Research Centre, University College Dublin, Dublin 4, Republic of Ireland
| | - Emma L Feeney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- UCD Institute for Food and Health, University College Dublin, Dublin 4, Republic of Ireland
| |
Collapse
|
5
|
Chen KL, Chou RH, Chang CC, Kuo CS, Wei JH, Huang PH, Lin SJ. The high-density lipoprotein cholesterol (HDL-C)-concentration-dependent association between anti-inflammatory capacity and sepsis: A single-center cross-sectional study. PLoS One 2024; 19:e0296863. [PMID: 38603717 PMCID: PMC11008828 DOI: 10.1371/journal.pone.0296863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/20/2023] [Indexed: 04/13/2024] Open
Abstract
INTRODUCTION Known to have pleiotropic functions, high-density lipoprotein (HDL) helps to regulate systemic inflammation during sepsis. As preserving HDL-C level is a promising therapeutic strategy for sepsis, the interaction between HDL and sepsis worth further investigation. This study aimed to determine the impact of sepsis on HDL's anti-inflammatory capacity and explore its correlations with disease severity and laboratory parameters. METHODS AND MATERIALS We enrolled 80 septic subjects admitted to the intensive care unit and 50 controls admitted for scheduled coronary angiography in this cross-sectional study. We used apolipoprotein-B depleted (apoB-depleted) plasma to measure the anti-inflammatory capacity of HDL-C. ApoB-depleted plasma's anti-inflammatory capacity is defined as its ability to suppress tumor necrosis factor-α-induced vascular cell adhesion molecule-1 (VCAM-1) expression in human umbilical-vein endothelial cells. A subgroup analysis was conducted to investigate in septic subjects according to disease severity. RESULTS ApoB-depleted plasma's anti-inflammatory capacity was reduced in septic subjects relative to controls (VCAM-1 mRNA fold change: 50.1% vs. 35.5%; p < 0.0001). The impairment was more pronounced in septic subjects with than in those without septic shock (55.8% vs. 45.3%, p = 0.0022). Both associations were rendered non-significant with the adjustment for the HDL-C level. In sepsis patients, VCAM-1 mRNA fold change correlated with the SOFA score (Spearman's r = 0.231, p = 0.039), lactate level (r = 0.297, p = 0.0074), HDL-C level (r = -0.370, p = 0.0007), and inflammatory markers (C-reactive protein level: r = 0.441, p <0.0001; white blood cell: r = 0.353, p = 0.0013). CONCLUSION ApoB-depleted plasma's anti-inflammatory capacity is reduced in sepsis patients and this association depends of HDL-C concentration. In sepsis patients, this capacity correlates with disease severity and inflammatory markers. These findings explain the prognostic role of the HDL-C level in sepsis and indirectly support the rationale for targeting HDL-C as sepsis treatment.
Collapse
Affiliation(s)
- Kai-Lee Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Chin Chang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Sung Kuo
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jih-Hua Wei
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Heart Center, Cheng-Hsin General Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Hu S, Zhu Y, Zhao X, Li R, Shao G, Gong D, Hu C, Liu H, Xu K, Liu C, Xu M, Zhao Z, Li T, Hu Z, Shao M, Liu J, Li X, Wu H, Li J, Xu Y. Hepatocytic lipocalin-2 controls HDL metabolism and atherosclerosis via Nedd4-1-SR-BI axis in mice. Dev Cell 2023; 58:2326-2337.e5. [PMID: 37863040 DOI: 10.1016/j.devcel.2023.09.007] [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: 02/28/2023] [Revised: 07/03/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023]
Abstract
High-density lipoprotein (HDL) metabolism is regulated by complex interplay between the scavenger receptor group B type 1 (SR-BI) and multiple signaling molecules in the liver. Here, we show that lipocalin-2 (Lcn2) is a key regulator of hepatic SR-BI, HDL metabolism, and atherosclerosis. Overexpression of human Lcn2 in hepatocytes attenuates the development of atherosclerosis via SR-BI in western-diet-fed Ldlr-/- mice, whereas hepatocyte-specific ablation of Lcn2 has the opposite effect. Mechanistically, hepatocyte Lcn2 improves HDL metabolism and alleviates atherogenesis by blocking Nedd4-1-mediated SR-BI ubiquitination at K500 and K508. The Lcn2-improved HDL metabolism is abolished in mice with hepatocyte-specific Nedd4-1 or SR-BI deletion and in SR-BI (K500A/K508A) mutation mice. This study identifies a regulatory axis from Lcn2 to HDL via blocking Nedd4-1-mediated SR-BI ubiquitination and demonstrates that hepatocyte Lcn2 may be a promising target to improve HDL metabolism to treat atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Shuwei Hu
- School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China; Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yingdong Zhu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Xiaojie Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Rui Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Guangze Shao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Dongxu Gong
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Chencheng Hu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Hongjun Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Kexin Xu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Chenxi Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Minghuan Xu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Tao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital Xizhimen South Street, West District, Beijing 100044, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Mengle Shao
- Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun- Liu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yanyong Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Pathology of School of Basic Medical Sciences, Frontier Innovation Center, Fudan University Shanghai, Shanghai 200032, China; Diabetes, Obesity and Metabolism Research Center, Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| |
Collapse
|
7
|
de Azúa-López ZR, Pezzotti MR, González-Díaz Á, Meilhac O, Ureña J, Amaya-Villar R, Castellano A, Varela LM. HDL anti-inflammatory function is impaired and associated with high SAA1 and low APOA4 levels in aneurysmal subarachnoid hemorrhage. J Cereb Blood Flow Metab 2023; 43:1919-1930. [PMID: 37357772 PMCID: PMC10676137 DOI: 10.1177/0271678x231184806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/07/2023] [Accepted: 06/02/2023] [Indexed: 06/27/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating disease with high morbidity and mortality rates. Within 24 hours after aSAH, monocytes are recruited and enter the subarachnoid space, where they mature into macrophages, increasing the inflammatory response and contributing, along with other factors, to delayed neurological dysfunction and poor outcomes. High-density lipoproteins (HDL) are lipid-protein complexes that exert anti-inflammatory effects but under pathological conditions undergo structural alterations that have been associated with loss of functionality. Plasma HDL were isolated from patients with aSAH and analyzed for their anti-inflammatory activity and protein composition. HDL isolated from patients lost the ability to prevent VCAM-1 expression in endothelial cells (HUVEC) and subsequent adhesion of THP-1 monocytes to the endothelium. Proteomic analysis showed that HDL particles from patients had an altered composition compared to those of healthy subjects. We confirmed by western blot that low levels of apolipoprotein A4 (APOA4) and high of serum amyloid A1 (SAA1) in HDL were associated with the lack of anti-inflammatory function observed in aSAH. Our results indicate that the study of HDL in the pathophysiology of aSAH is needed, and functional HDL supplementation could be considered a novel therapeutic approach to the treatment of the inflammatory response after aSAH.
Collapse
Affiliation(s)
- Zaida Ruiz de Azúa-López
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Unidad de Cuidados Intensivos, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - M Rosa Pezzotti
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Ángela González-Díaz
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), Saint-Pierre de La Réunion, France
- CHU de La Réunion, Saint-Pierre de la Réunion, France
| | - Juan Ureña
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Rosario Amaya-Villar
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Unidad de Cuidados Intensivos, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Antonio Castellano
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Lourdes M Varela
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
8
|
Nieddu G, Formato M, Lepedda AJ. Searching for Atherosclerosis Biomarkers by Proteomics: A Focus on Lesion Pathogenesis and Vulnerability. Int J Mol Sci 2023; 24:15175. [PMID: 37894856 PMCID: PMC10607641 DOI: 10.3390/ijms242015175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Plaque rupture and thrombosis are the most important clinical complications in the pathogenesis of stroke, coronary arteries, and peripheral vascular diseases. The identification of early biomarkers of plaque presence and susceptibility to ulceration could be of primary importance in preventing such life-threatening events. With the improvement of proteomic tools, large-scale technologies have been proven valuable in attempting to unravel pathways of atherosclerotic degeneration and identifying new circulating markers to be utilized either as early diagnostic traits or as targets for new drug therapies. To address these issues, different matrices of human origin, such as vascular cells, arterial tissues, plasma, and urine, have been investigated. Besides, proteomics was also applied to experimental atherosclerosis in order to unveil significant insights into the mechanisms influencing atherogenesis. This narrative review provides an overview of the last twenty years of omics applications to the study of atherogenesis and lesion vulnerability, with particular emphasis on lipoproteomics and vascular tissue proteomics. Major issues of tissue analyses, such as plaque complexity, sampling, availability, choice of proper controls, and lipoproteins purification, will be raised, and future directions will be addressed.
Collapse
Affiliation(s)
| | | | - Antonio Junior Lepedda
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (G.N.); (M.F.); Antonio Junior Lepedda (A.J.L.)
| |
Collapse
|
9
|
Sergi D, Zauli E, Tisato V, Secchiero P, Zauli G, Cervellati C. Lipids at the Nexus between Cerebrovascular Disease and Vascular Dementia: The Impact of HDL-Cholesterol and Ceramides. Int J Mol Sci 2023; 24:ijms24054403. [PMID: 36901834 PMCID: PMC10002119 DOI: 10.3390/ijms24054403] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cerebrovascular diseases and the subsequent brain hypoperfusion are at the basis of vascular dementia. Dyslipidemia, marked by an increase in circulating levels of triglycerides and LDL-cholesterol and a parallel decrease in HDL-cholesterol, in turn, is pivotal in promoting atherosclerosis which represents a common feature of cardiovascular and cerebrovascular diseases. In this regard, HDL-cholesterol has traditionally been considered as being protective from a cardiovascular and a cerebrovascular prospective. However, emerging evidence suggests that their quality and functionality play a more prominent role than their circulating levels in shaping cardiovascular health and possibly cognitive function. Furthermore, the quality of lipids embedded in circulating lipoproteins represents another key discriminant in modulating cardiovascular disease, with ceramides being proposed as a novel risk factor for atherosclerosis. This review highlights the role of HDL lipoprotein and ceramides in cerebrovascular diseases and the repercussion on vascular dementia. Additionally, the manuscript provides an up-to-date picture of the impact of saturated and omega-3 fatty acids on HDL circulating levels, functionality and ceramide metabolism.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| | - Enrico Zauli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Zauli
- King Khaled Eye Specialistic Hospital, Riyadh 11462, Saudi Arabia
| | - Carlo Cervellati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
10
|
Expanding the Molecular Disturbances of Lipoproteins in Cardiometabolic Diseases: Lessons from Lipidomics. Diagnostics (Basel) 2023; 13:diagnostics13040721. [PMID: 36832218 PMCID: PMC9954993 DOI: 10.3390/diagnostics13040721] [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: 01/02/2023] [Revised: 01/28/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The increasing global burden of cardiometabolic diseases highlights the urgent clinical need for better personalized prediction and intervention strategies. Early diagnosis and prevention could greatly reduce the enormous socio-economic burden posed by these states. Plasma lipids including total cholesterol, triglycerides, HDL-C, and LDL-C have been at the center stage of the prediction and prevention strategies for cardiovascular disease; however, the bulk of cardiovascular disease events cannot be explained sufficiently by these lipid parameters. The shift from traditional serum lipid measurements that are poorly descriptive of the total serum lipidomic profile to comprehensive lipid profiling is an urgent need, since a wealth of metabolic information is currently underutilized in the clinical setting. The tremendous advances in the field of lipidomics in the last two decades has facilitated the research efforts to unravel the lipid dysregulation in cardiometabolic diseases, enabling the understanding of the underlying pathophysiological mechanisms and identification of predictive biomarkers beyond traditional lipids. This review presents an overview of the application of lipidomics in the study of serum lipoproteins in cardiometabolic diseases. Integrating the emerging multiomics with lipidomics holds great potential in moving toward this goal.
Collapse
|
11
|
Avdeeva AS. Inflammatory markers in rheumatic diseases. RHEUMATOLOGY SCIENCE AND PRACTICE 2022. [DOI: 10.47360/1995-4484-2022-561-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immune-mediated rheumatic diseases (IMRDs) are a broad group of pathological conditions based on impaired immunological tolerance to one’s own tissues leading to inflammation and irreversible organ damage. Laboratory diagnosis of IMRDs includes a wide range of biomarkers (autoantibodies, acute phase proteins, cytokines, markers of endothelial damage, components of the complement system, immunoglobulins, cryoglobulins, lymphocyte subpopulations, indicators of bone metabolism, apoptosis markers, genetic markers, etc). One of the leading aspects of laboratory diagnosis of IMRDs is the study of the level of inflammation markers in the blood (erythrocyte sedimentation rate, C-reactive protein (CRP), serum amyloid protein (CAA), ferritin, procalcitonin, apolipoprotein AI, calprotectin, etc). The analysis of inflammation markers makes it possible to assess the disease activity, the nature of the progression and the prognosis of the outcomes of a chronic inflammatory process, as well as the effectiveness of the therapy. The review presents the latest data on the role of the most frequently studied inflammatory markers such as CRP, CAA and ferritin.
Collapse
|
12
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
13
|
Uremic mouse model to study vascular calcification and "inflamm-aging". J Mol Med (Berl) 2022; 100:1321-1330. [PMID: 35916902 PMCID: PMC9402761 DOI: 10.1007/s00109-022-02234-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/19/2022] [Accepted: 07/05/2022] [Indexed: 11/05/2022]
Abstract
Calcification and chronic inflammation of the vascular wall is a high-risk factor for cardiovascular mortality, especially in patients with chronic uremia. For the reduction or prevention of rapid disease progression, no specific treatment options are currently available. This study aimed to evaluate an adenine-based uremic mouse model for studying medial vessel calcification and senescence-associated secretory phenotype (SASP) changes of aortic tissue to unravel molecular pathogenesis and provide a model for therapy testing. The dietary adenine administration induced a stable and similar degree of chronic uremia in DBA2/N mice with an increase of uremia blood markers such as blood urea nitrogen, calcium, creatinine, alkaline phosphatase, and parathyroid hormone. Also, renal fibrosis and crystal deposits were detected upon adenine feeding. The uremic condition is related to a moderate to severe medial vessel calcification and subsequent elastin disorganization. In addition, expression of osteogenic markers as Bmp-2 and its transcription factor Sox-9 as well as p21 as senescence marker were increased in uremic mice compared to controls. Pro-inflammatory uremic proteins such as serum amyloid A, interleukin (Il)-1β, and Il-6 increased. This novel model of chronic uremia provides a simple method for investigation of signaling pathways in vascular inflammation and calcification and therefore offers an experimental basis for the development of potential therapeutic intervention studies.
Collapse
|
14
|
Karabacak M, Uysal BA, Turkdogan AK. Alteration in serum oxidative stress balance in patients with different circulating high-density lipoprotein cholesterol levels. Rev Port Cardiol 2022; 41:833-839. [DOI: 10.1016/j.repc.2021.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 11/29/2022] Open
|
15
|
Romo EZ, Zivkovic AM. Glycosylation of HDL-Associated Proteins and Its Implications in Cardiovascular Disease Diagnosis, Metabolism and Function. Front Cardiovasc Med 2022; 9:928566. [PMID: 35694676 PMCID: PMC9184513 DOI: 10.3389/fcvm.2022.928566] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/09/2022] [Indexed: 01/09/2023] Open
Abstract
High-density lipoprotein (HDL) particles, long known for their critical role in the prevention of cardiovascular disease (CVD), were recently identified to carry a wide array of glycosylated proteins, and the importance of this glycosylation in the structure, function and metabolism of HDL are starting to emerge. Early studies have demonstrated differential glycosylation of HDL-associated proteins in various pathological states, which may be key to understanding their etiological role in these diseases and may be important for diagnostic development. Given the vast array and specificity of glycosylation pathways, the study of HDL-associated glycosylation has the potential to uncover novel mechanisms and biomarkers of CVD. To date, no large studies examining the relationships between HDL glycosylation profiles and cardiovascular outcomes have been performed. However, small pilot studies provide promising preliminary evidence that such a relationship may exist. In this review article we discuss the current state of the evidence on the glycosylation of HDL-associated proteins, the potential for HDL glycosylation profiling in CVD diagnostics, how glycosylation affects HDL function, and the potential for modifying the glycosylation of HDL-associated proteins to confer therapeutic value.
Collapse
|
16
|
Gibson CM, Kazmi SHA, Korjian S, Chi G, Phillips AT, Montazerin SM, Duffy D, Zheng B, Heise M, Liss C, Deckelbaum LI, Wright SD, Gille A. CSL112 (Apolipoprotein A-I [Human]) Strongly Enhances Plasma Apoa-I and Cholesterol Efflux Capacity in Post-Acute Myocardial Infarction Patients: A PK/PD Substudy of the AEGIS-I Trial. J Cardiovasc Pharmacol Ther 2022; 27:10742484221121507. [DOI: 10.1177/10742484221121507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Introduction: Cholesterol efflux capacity (CEC) is impaired following acute myocardial infarction (AMI). CSL112 is an intravenous preparation of human plasma-derived apoA-I formulated with phosphatidylcholine (PC). CSL112 is intended to improve CEC and thereby prevent early recurrent cardiovascular events following AMI. AEGIS-I (ApoA-I Event Reducing in Ischemic Syndromes I) was a multicenter, randomized, double-blind, placebo-controlled, dose-ranging phase 2b study, designed to evaluate the hepatic and renal safety of CSL112. Here, we report an analysis of a pharmacokinetic (PK) and pharmacodynamic (PD) substudy of AEGIS-I. Methods: AMI patients were stratified by renal function and randomized 3:3:2 to 4, weekly, 2-hour infusions of low- and high-dose (2 g and 6 g) CSL112, or placebo. PK/PD assessments included plasma concentrations of apoA-I and PC, and measures of total and ABCA1-dependent CEC, as well as lipids/lipoproteins including high density lipoprotein cholesterol (HDL-C), non-HDL-C, low density lipoprotein cholesterol (LDL-C), ApoB, and triglycerides. Inflammatory and cardio-metabolic biomarkers were also evaluated. Results: The substudy included 63 subjects from AEGIS-I. CSL112 infusions resulted in rapid, dose-dependent increases in baseline corrected apoA-I and PC, which peaked at the end of the infusion (Tmax ≈ 2 hours). Similarly, there was a dose-dependent elevation in both total CEC and ABCA1-mediated CEC. Mild renal impairment did not affect the PK or PD of CSL112. CSL112 administration was also associated with an increase in plasma levels of HDL-C but not non-HDL-C, LDL-C, apoB, or triglycerides. No dose-effects on inflammatory or cardio-metabolic biomarkers were observed. Conclusion: Among patients with AMI, impaired CEC was rapidly elevated by CSL112 infusions in a dose-dependent fashion, along with an increase in apoA-I plasma concentrations. Findings from the current sub-study of the AEGIS-I support a potential atheroprotective benefit of CSL112 for AMI patients.
Collapse
Affiliation(s)
- C. Michael Gibson
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Syed Hassan A. Kazmi
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Serge Korjian
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gerald Chi
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Adam T. Phillips
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sahar Memar Montazerin
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Bo Zheng
- CSL Behring, King of Prussia, PA, USA
| | | | | | | | | | | |
Collapse
|
17
|
Souza Junior DR, Silva ARM, Rosa-Fernandes L, Reis LR, Alexandria G, Bhosale SD, Ghilardi FDR, Dalçóquio TF, Bertolin AJ, Nicolau JC, Marinho CRF, Wrenger C, Larsen MR, Siciliano RF, Di Mascio P, Palmisano G, Ronsein GE. HDL proteome remodeling associates with COVID-19 severity. J Clin Lipidol 2021; 15:796-804. [PMID: 34802985 PMCID: PMC8557113 DOI: 10.1016/j.jacl.2021.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 10/25/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Besides the well-accepted role in lipid metabolism, high-density lipoprotein (HDL) also seems to participate in host immune response against infectious diseases. OBJECTIVE We used a quantitative proteomic approach to test the hypothesis that alterations in HDL proteome associate with severity of Coronavirus disease 2019 (COVID-19). METHODS Based on clinical criteria, subjects (n=41) diagnosed with COVID-19 were divided into two groups: a group of subjects presenting mild symptoms and a second group displaying severe symptoms and requiring hospitalization. Using a proteomic approach, we quantified the levels of 29 proteins in HDL particles derived from these subjects. RESULTS We showed that the levels of serum amyloid A 1 and 2 (SAA1 and SAA2, respectively), pulmonary surfactant-associated protein B (SFTPB), apolipoprotein F (APOF), and inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4) were increased by more than 50% in hospitalized patients, independently of sex, HDL-C or triglycerides when comparing with subjects presenting only mild symptoms. Altered HDL proteins were able to classify COVID-19 subjects according to the severity of the disease (error rate 4.9%). Moreover, apolipoprotein M (APOM) in HDL was inversely associated with odds of death due to COVID-19 complications (odds ratio [OR] per 1-SD increase in APOM was 0.27, with 95% confidence interval [CI] of 0.07 to 0.72, P=0.007). CONCLUSION Our results point to a profound inflammatory remodeling of HDL proteome tracking with severity of COVID-19 infection. They also raise the possibility that HDL particles could play an important role in infectious diseases.
Collapse
Affiliation(s)
| | | | - Livia Rosa-Fernandes
- Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Lorenna Rocha Reis
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil
| | - Gabrielly Alexandria
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil
| | - Santosh D Bhosale
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | | | | | - José Carlos Nicolau
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Claudio R F Marinho
- Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Rinaldo Focaccia Siciliano
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil; Division of Infectious and Parasitic Diseases, University of São Paulo Medical School, São Paulo, Brazil
| | - Paolo Di Mascio
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil.
| | - Graziella Eliza Ronsein
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
18
|
Kochan Z, Szupryczynska N, Malgorzewicz S, Karbowska J. Dietary Lipids and Dyslipidemia in Chronic Kidney Disease. Nutrients 2021; 13:3138. [PMID: 34579015 PMCID: PMC8472557 DOI: 10.3390/nu13093138] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
The progression of chronic kidney disease (CKD) leads to altered lipid metabolism. CKD patients exhibit high blood triglyceride (TG) levels, reduced concentrations and functionality of high-density lipoproteins (HDL), and elevated levels of atherogenic small, dense, low-density lipoproteins (sdLDL). Disorders of lipid metabolism and other metabolic disturbances place CKD patients at high risk for cardiovascular disease (CVD). Extensive evidence supports the cardioprotective effects of unsaturated fatty acids, including their beneficial effect on serum cholesterol and TG levels. Dietary lipids might therefore be especially important in the nutritional management of CKD. We review current dietary recommendations for fat intake by CKD patients and suggest potential nutritional interventions by emphasizing dietary lipids that might improve the blood lipid profile and reduce cardiovascular risk in CKD.
Collapse
Affiliation(s)
- Zdzislaw Kochan
- Laboratory of Nutritional Biochemistry, Department of Clinical Nutrition, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland; (Z.K.); (N.S.)
| | - Natalia Szupryczynska
- Laboratory of Nutritional Biochemistry, Department of Clinical Nutrition, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland; (Z.K.); (N.S.)
| | - Sylwia Malgorzewicz
- Department of Clinical Nutrition, Division of Clinical Nutrition and Dietetics, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Joanna Karbowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
19
|
Ehlting C, Wolf SD, Bode JG. Acute-phase protein synthesis: a key feature of innate immune functions of the liver. Biol Chem 2021; 402:1129-1145. [PMID: 34323429 DOI: 10.1515/hsz-2021-0209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023]
Abstract
The expression of acute-phase proteins (APP's) maintains homeostasis and tissue repair, but also represents a central component of the organism's defense strategy, especially in the context of innate immunity. Accordingly, an inflammatory response is accompanied by significant changes in the serum protein composition, an aspect that is also used diagnostically. As the main site of APP synthesis the liver is constantly exposed to antigens or pathogens via blood flow, but also to systemic inflammatory signals originating either from the splanchnic area or from the circulation. Under both homeostatic and acute-phase response (APR) conditions the composition of APP's is determined by the pattern of regulatory mediators derived from the systemic circulation or from local cell populations, especially liver macrophages. The key regulators mentioned here most frequently are IL-1β, IL-6 and TNF-α. In addition to a variety of molecular mediators described mainly on the basis of in vitro studies, recent data emphasize the in vivo relevance of cellular key effectors as well as molecular key mediators and protein modifications for the regulation and function of APP's. These are aspects, on which the present review is primarily focused.
Collapse
Affiliation(s)
- Christian Ehlting
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Hospital of the Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Stephanie D Wolf
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Hospital of the Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Johannes G Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Hospital of the Heinrich-Heine-University, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
20
|
Kaseda R, Hosojima M, Kuwahara S, Kabasawa H, Aoki H, Higuchi Y, Kon V, Narita I, Saito A. Rice Endosperm Protein Improves the Anti-Inflammatory Effects of High-Density Lipoprotein and Produces Lower Atherosclerotic Lesion Accelerated by the Renal Mass Reduction than Casein in a Mouse Model. J Am Coll Nutr 2021; 41:668-678. [PMID: 34424818 DOI: 10.1080/07315724.2021.1950584] [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: 10/20/2022]
Abstract
Chronic kidney disease (CKD) impairs the anti-inflammatory effects of high-density lipoprotein (HDL) and increases cardiovascular mortality. Though the potential role of dietary interventions to manage HDL is well studied, the clinical trials aimed to increase HDL levels have failed to reduce cardiovascular risk, rendering HDL function to be explored as a more relevant clinical parameter. This study investigates the effects of rice endosperm protein (REP), a plant-based protein, on the anti-inflammatory properties of HDL and renal injury-driven atherosclerosis in comparison with casein, an animal protein. Ten-week-old apolipoprotein E-deficient hyperlipidemic mice underwent uninephrectomy. The mice (n = 6 each) were pair-fed a normal casein-based diet or a REP-based diet (both with 20.0% protein content) for seven weeks. Atherosclerotic lesions were detected by en face Sudan IV staining of the aorta. The number and sizes of the atherosclerotic lesions were significantly lower in the REP-based diet-fed group than the casein-based diet-fed group (p = 0.038). However, the REP-based diet neither elicited an ameliorative effect on kidney function or histology nor impacted the cholesterol profiles. Furthermore, HDL from the REP-based diet-fed mice significantly suppressed the inflammatory cytokine response of human umbilical vein endothelial cells than that from the casein-based diet-fed mice (MCP-1, p = 0.010; IL-6, p = 0.011; IL-1β, p = 0.028). The REP-based diet has a higher potential to lessen the atherosclerotic lesions accelerated by renal mass reduction than a casein-based diet, which could be associated with the anti-inflammatory effects of HDL.
Collapse
Affiliation(s)
- Ryohei Kaseda
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Michihiro Hosojima
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Shoji Kuwahara
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Hideyuki Kabasawa
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Hiroyuki Aoki
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Yuki Higuchi
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan.,Rice Research Center, Kameda Seika Co. Ltd, Niigata, Japan
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Akihiko Saito
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| |
Collapse
|
21
|
Noels H, Lehrke M, Vanholder R, Jankowski J. Lipoproteins and fatty acids in chronic kidney disease: molecular and metabolic alterations. Nat Rev Nephrol 2021; 17:528-542. [PMID: 33972752 DOI: 10.1038/s41581-021-00423-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) induces modifications in lipid and lipoprotein metabolism and homeostasis. These modifications can promote, modulate and/or accelerate CKD and secondary cardiovascular disease (CVD). Lipid and lipoprotein abnormalities - involving triglyceride-rich lipoproteins, LDL and/or HDL - not only involve changes in concentration but also changes in molecular structure, including protein composition, incorporation of small molecules and post-translational modifications. These alterations modify the function of lipoproteins and can trigger pro-inflammatory and pro-atherogenic processes, as well as oxidative stress. Serum fatty acid levels are also often altered in patients with CKD and lead to changes in fatty acid metabolism - a key process in intracellular energy production - that induce mitochondrial dysfunction and cellular damage. These fatty acid changes might not only have a negative impact on the heart, but also contribute to the progression of kidney damage. The presence of these lipoprotein alterations within a biological environment characterized by increased inflammation and oxidative stress, as well as the competing risk of non-atherosclerotic cardiovascular death as kidney function declines, has important therapeutic implications. Additional research is needed to clarify the pathophysiological link between lipid and lipoprotein modifications, and kidney dysfunction, as well as the genesis and/or progression of CVD in patients with kidney disease.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Michael Lehrke
- Department of Internal Medicine I, RWTH Aachen University, University Hospital, Aachen, Germany
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany.
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
22
|
Kon V, Yang HC, Smith LE, Vickers KC, Linton MF. High-Density Lipoproteins in Kidney Disease. Int J Mol Sci 2021; 22:ijms22158201. [PMID: 34360965 PMCID: PMC8348850 DOI: 10.3390/ijms22158201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Decades of epidemiological studies have established the strong inverse relationship between high-density lipoprotein (HDL)-cholesterol concentration and cardiovascular disease. Recent evidence suggests that HDL particle functions, including anti-inflammatory and antioxidant functions, and cholesterol efflux capacity may be more strongly associated with cardiovascular disease protection than HDL cholesterol concentration. These HDL functions are also relevant in non-cardiovascular diseases, including acute and chronic kidney disease. This review examines our current understanding of the kidneys’ role in HDL metabolism and homeostasis, and the effect of kidney disease on HDL composition and functionality. Additionally, the roles of HDL particles, proteins, and small RNA cargo on kidney cell function and on the development and progression of both acute and chronic kidney disease are examined. The effect of HDL protein modification by reactive dicarbonyls, including malondialdehyde and isolevuglandin, which form adducts with apolipoprotein A-I and impair proper HDL function in kidney disease, is also explored. Finally, the potential to develop targeted therapies that increase HDL concentration or functionality to improve acute or chronic kidney disease outcomes is discussed.
Collapse
Affiliation(s)
- Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (V.K.); (H.-C.Y.)
| | - Hai-Chun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (V.K.); (H.-C.Y.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Loren E. Smith
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Kasey C. Vickers
- Atherosclerosis Research Unit, Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - MacRae F. Linton
- Atherosclerosis Research Unit, Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
23
|
Wu B, Niu Z, Hu F. Study on Risk Factors of Peripheral Neuropathy in Type 2 Diabetes Mellitus and Establishment of Prediction Model. Diabetes Metab J 2021; 45:526-538. [PMID: 34352988 PMCID: PMC8369209 DOI: 10.4093/dmj.2020.0100] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is one of the most serious complications of type 2 diabetes mellitus (T2DM). DPN increases the risk of ulcers, foot infections, and noninvasive amputations, ultimately leading to long-term disability. METHODS Seven hundred patients with T2DM were investigated from 2013 to 2017 in the Sanlin community by obtaining basic data from the electronic medical record system (EMRS). From September 2018 to July 2019, 681 patients (19 missing) were investigated using a questionnaire, physical examination, biochemical index test, and follow-up Toronto clinical scoring system (TCSS) test. Patients with a TCSS score ≥6 points were diagnosed with DPN. After removing missing values, 612 patients were divided into groups in a 3:1 ratio for external validation. Using different Lasso analyses (misclassification error, mean squared error, -2log-likelihood, and area under curve) and a logistic regression analysis of the training set, models A, B, C, and D were established. The receiver operating characteristic (ROC) curve, calibration plot, dynamic component analysis (DCA) measurements, net classification improvement (NRI) and integrated discrimination improvement (IDI) were used to validate discrimination and clinical practicality of the model. RESULTS Through data analysis, model A (containing four factors), model B (containing five factors), model C (containing seven factors), and model D (containing seven factors) were built. After calibration, ROC curve, DCA, NRI and IDI, models C and D exhibited better accuracy and greater predictive power. CONCLUSION Four prediction models were established to assist with the early screening of DPN in patients with T2DM. The influencing factors in model C and D are more important factors for patients with T2DM diagnosed with DPN.
Collapse
Affiliation(s)
- Birong Wu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheyun Niu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fan Hu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Corresponding author: Fan Hu https://orcid.org/0000-0002-7929-6953 School of Public Health, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China E-mail:
| |
Collapse
|
24
|
Jia C, Anderson JLC, Gruppen EG, Lei Y, Bakker SJL, Dullaart RPF, Tietge UJF. High-Density Lipoprotein Anti-Inflammatory Capacity and Incident Cardiovascular Events. Circulation 2021; 143:1935-1945. [PMID: 33840204 DOI: 10.1161/circulationaha.120.050808] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The role of high-density lipoprotein (HDL) function in cardiovascular disease represents an important emerging concept. The present study investigated whether HDL anti-inflammatory capacity is prospectively associated with first cardiovascular events in the general population. METHODS HDL anti-inflammatory capacity was determined as its ability to suppress TNFα (tumor necrosis factor α)-induced VCAM-1 (vascular cell adhesion molecule-1) mRNA expression in endothelial cells in vitro (results expressed as achieved percent reduction by individual HDL related to the maximum TNFα effect with no HDL present). In a nested case-control design of the PREVEND (Prevention of Renal and Vascular End Stage Disease) study, 369 cases experiencing a first cardiovascular event (combined end point of death from cardiovascular causes, ischemic heart disease, nonfatal myocardial infarction, and coronary revascularization) during a median of 10.5 years of follow-up were identified and individually matched to 369 controls with respect to age, sex, smoking status, and HDL cholesterol. Baseline samples were available in 340 cases and 340 matched controls. RESULTS HDL anti-inflammatory capacity was not correlated with HDL cholesterol or hsCRP (high-sensitivity C-reactive protein). HDL anti-inflammatory capacity was significantly lower in cases compared with controls (31.6% [15.7-44.2] versus 27.0% [7.4-36.1]; P<0.001) and was inversely associated with incident CVD in a fully adjusted model (odds ratio [OR] per 1 SD, 0.74 [CI, 0.61-0.90]; P=0.002). Furthermore, this association was approximately similar with all individual components of the cardiovascular disease end point. The HDL anti-inflammatory was not correlated with cholesterol efflux capacity (r=-0.02; P>0.05). When combining these 2 HDL function metrics in 1 model, both were significantly and independently associated with incident cardiovascular disease in a fully adjusted model (efflux: OR per 1 SD, 0.74; P=0.002; anti-inflammatory capacity: OR per 1 SD, 0.66; P<0.001). Adding HDL anti-inflammatory capacity improved risk prediction by the Framingham risk score, with a model likelihood-ratio statistic increase from 10.50 to 20.40 (P=0.002). CONCLUSIONS The HDL anti-inflammatory capacity, reflecting vascular protection against key steps in atherogenesis, was inversely associated with incident cardiovascular events in a general population cohort, independent of HDL cholesterol and HDL cholesterol efflux capacity. Adding HDL anti-inflammatory capacity to the Framingham risk score improves risk prediction.
Collapse
Affiliation(s)
- Congzhuo Jia
- Department of Pediatrics (C.J., J.L.C.A., U.J.F.T.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (C.J., Y.L., U.J.F.T.)
| | - Josephine L C Anderson
- Department of Pediatrics (C.J., J.L.C.A., U.J.F.T.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eke G Gruppen
- Department of Endocrinology, (E.G.G., R.P.F.D.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Nephrology (E.G.G., S.J.L.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (C.J., Y.L., U.J.F.T.)
| | - Stephan J L Bakker
- Department of Nephrology (E.G.G., S.J.L.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, (E.G.G., R.P.F.D.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics (C.J., J.L.C.A., U.J.F.T.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (C.J., Y.L., U.J.F.T.)
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden (U.J.F.T.)
| |
Collapse
|
25
|
Coimbra S, Reis F, Valente MJ, Rocha S, Catarino C, Rocha-Pereira P, Sameiro-Faria M, Bronze-da-Rocha E, Belo L, Santos-Silva A. Subpopulations of High-Density Lipoprotein: Friends or Foes in Cardiovascular Disease Risk in Chronic Kidney Disease? Biomedicines 2021; 9:554. [PMID: 34065648 PMCID: PMC8157071 DOI: 10.3390/biomedicines9050554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/23/2022] Open
Abstract
Dyslipidemia is a major traditional risk factor for cardiovascular disease (CVD) in chronic kidney disease (CKD) patients, although the altered lipid profile does not explain the number and severity of CVD events. High-density lipoprotein (HDL) is a heterogeneous (size, composition, and functionality) population of particles with different atherogenic or atheroprotective properties. HDL-cholesterol concentrations per se may not entirely reflect a beneficial or a risk profile for CVD. Large HDL in CKD patients may have a unique proteome and lipid composition, impairing their cholesterol efflux capacity. This lack of HDL functionality may contribute to the paradoxical coexistence of increased large HDL and enhanced risk for CVD events. Moreover, CKD is associated with inflammation, oxidative stress, diabetes, and/or hypertension that are able to interfere with the anti-inflammatory, antioxidative, and antithrombotic properties of HDL subpopulations. How these changes interfere with HDL functions in CKD is still poorly understood. Further studies are warranted to fully clarify if different HDL subpopulations present different functionalities and/or atheroprotective effects. To achieve this goal, the standardization of techniques would be valuable.
Collapse
Affiliation(s)
- Susana Coimbra
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), 4585-116 Gandra-Paredes, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics, & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Maria João Valente
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
| | - Susana Rocha
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Cristina Catarino
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
| | - Petronila Rocha-Pereira
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
- Chemistry Department, University of Beira Interior, 6201-001 Covilhã, Portugal
| | - Maria Sameiro-Faria
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
- Hemodialysis Clinic Hospital Agostinho Ribeiro, 4610-106 Felgueiras, Portugal
| | - Elsa Bronze-da-Rocha
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
| | - Luís Belo
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
| | - Alice Santos-Silva
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.J.V.); (C.C.); (P.R.-P.); (M.S.-F.); (E.B.-d.-R.); (L.B.); (A.S.-S.)
| |
Collapse
|
26
|
High-Density Lipoproteins and the Kidney. Cells 2021; 10:cells10040764. [PMID: 33807271 PMCID: PMC8065870 DOI: 10.3390/cells10040764] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Dyslipidemia is a typical trait of patients with chronic kidney disease (CKD) and it is typically characterized by reduced high-density lipoprotein (HDL)-cholesterol(c) levels. The low HDL-c concentration is the only lipid alteration associated with the progression of renal disease in mild-to-moderate CKD patients. Plasma HDL levels are not only reduced but also characterized by alterations in composition and structure, which are responsible for the loss of atheroprotective functions, like the ability to promote cholesterol efflux from peripheral cells and antioxidant and anti-inflammatory proprieties. The interconnection between HDL and renal function is confirmed by the fact that genetic HDL defects can lead to kidney disease; in fact, mutations in apoA-I, apoE, apoL, and lecithin–cholesterol acyltransferase (LCAT) are associated with the development of renal damage. Genetic LCAT deficiency is the most emblematic case and represents a unique tool to evaluate the impact of alterations in the HDL system on the progression of renal disease. Lipid abnormalities detected in LCAT-deficient carriers mirror the ones observed in CKD patients, which indeed present an acquired LCAT deficiency. In this context, circulating LCAT levels predict CKD progression in individuals at early stages of renal dysfunction and in the general population. This review summarizes the main alterations of HDL in CKD, focusing on the latest update of acquired and genetic LCAT defects associated with the progression of renal disease.
Collapse
|
27
|
Cohen G. Effect of High-Density Lipoprotein from Healthy Subjects and Chronic Kidney Disease Patients on the CD14 Expression on Polymorphonuclear Leukocytes. Int J Mol Sci 2021; 22:ijms22062830. [PMID: 33799511 PMCID: PMC7998954 DOI: 10.3390/ijms22062830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/26/2021] [Accepted: 03/08/2021] [Indexed: 01/22/2023] Open
Abstract
In uremic patients, high-density lipoprotein (HDL) loses its anti-inflammatory features and can even become pro-inflammatory due to an altered protein composition. In chronic kidney disease (CKD), impaired functions of polymorphonuclear leukocytes (PMNLs) contribute to inflammation and an increased risk of cardiovascular disease. This study investigated the effect of HDL from CKD and hemodialysis (HD) patients on the CD14 expression on PMNLs. HDL was isolated using a one-step density gradient centrifugation. Isolation of PMNLs was carried out by discontinuous Ficoll-Hypaque density gradient centrifugation. CD14 surface expression was quantified by flow cytometry. The activity of the small GTPase Rac1 was determined by means of an activation pull-down assay. HDL increased the CD14 surface expression on PMNLs. This effect was more pronounced for HDL isolated from uremic patients. The acute phase protein serum amyloid A (SAA) caused higher CD14 expression, while SAA as part of an HDL particle did not. Lipid raft disruption with methyl-β-cyclodextrin led to a reduced CD14 expression in the absence and presence of HDL. HDL from healthy subjects but not from HD patients decreased the activity of Rac1. Considering the known anti-inflammatory effects of HDL, the finding that even HDL from healthy subjects increased the CD14 expression was unexpected. The pathophysiological relevance of this result needs further investigation.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
28
|
Sorić Hosman I, Kos I, Lamot L. Serum Amyloid A in Inflammatory Rheumatic Diseases: A Compendious Review of a Renowned Biomarker. Front Immunol 2021; 11:631299. [PMID: 33679725 PMCID: PMC7933664 DOI: 10.3389/fimmu.2020.631299] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein with a significant importance for patients with inflammatory rheumatic diseases (IRD). The central role of SAA in pathogenesis of IRD has been confirmed by recent discoveries, including its involvement in the activation of the inflammasome cascade and recruitment of interleukin 17 producing T helper cells. Clinical utility of SAA in IRD was originally evaluated nearly half a century ago. From the first findings, it was clear that SAA could be used for evaluating disease severity and monitoring disease activity in patients with rheumatoid arthritis and secondary amyloidosis. However, cost-effective and more easily applicable markers, such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), overwhelmed its use in clinical practice. In the light of emerging evidences, SAA has been discerned as a more sensitive biomarker in a wide spectrum of IRD, especially in case of subclinical inflammation. Furthermore, a growing number of studies are confirming the advantages of SAA over many other biomarkers in predicting and monitoring response to biological immunotherapy in IRD patients. Arising scientific discoveries regarding the role of SAA, as well as delineating SAA and its isoforms as the most sensitive biomarkers in various IRD by recently developing proteomic techniques are encouraging the revival of its clinical use. Finally, the most recent findings have shown that SAA is a biomarker of severe Coronavirus disease 2019 (COVID-19). The aim of this review is to discuss the SAA-involving immune system network with emphasis on mechanisms relevant for IRD, as well as usefulness of SAA as a biomarker in various IRD. Therefore, over a hundred original papers were collected through an extensive PubMed and Scopus databases search. These recently arising insights will hopefully lead to a better management of IRD patients and might even inspire the development of new therapeutic strategies with SAA as a target.
Collapse
Affiliation(s)
- Iva Sorić Hosman
- Department of Pediatrics, Zadar General Hospital, Zadar, Croatia
| | - Ivanka Kos
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Lovro Lamot
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia.,Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Serum amyloid A (SAA) is a highly sensitive acute phase reactant that has been linked to a number of chronic inflammatory diseases. During a systemic inflammatory response, liver-derived SAA is primarily found on high-density lipoprotein (HDL). The purpose of this review is to discuss recent literature addressing the pathophysiological functions of SAA and the significance of its association with HDL. RECENT FINDINGS Studies in gene-targeted mice establish that SAA contributes to atherosclerosis and some metastatic cancers. Accumulating evidence indicates that the lipidation state of SAA profoundly affects its bioactivities, with lipid-poor, but not HDL-associated, SAA capable of inducing inflammatory responses in vitro and in vivo. Factors that modulate the equilibrium between lipid-free and HDL-associated SAA have been identified. HDL may serve to limit SAA's bioactivities in vivo. Understanding the factors leading to the release of systemic SAA from HDL may provide insights into chronic disease mechanisms.
Collapse
Affiliation(s)
- Nancy R Webb
- Department of Pharmacology and Nutritional Sciences, Saha Cardiovascular Research Center, and Barnstable Brown Diabetes Center, University of Kentucky, 553 Wethington Building, 900 South Limestone, Lexington, KY, 40536-0200, USA.
| |
Collapse
|
30
|
Gomes Kjerulf D, Wang S, Omer M, Pathak A, Subramanian S, Han CY, Tang C, den Hartigh LJ, Shao B, Chait A. Glycation of HDL blunts its anti-inflammatory and cholesterol efflux capacities in vitro, but has no effect in poorly controlled type 1 diabetes subjects. J Diabetes Complications 2020; 34:107693. [PMID: 32900591 PMCID: PMC7669727 DOI: 10.1016/j.jdiacomp.2020.107693] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND High-density lipoproteins (HDL) modified by glycation have been reported to be dysfunctional. Little is known regarding the anti-inflammatory effects on adipocytes of glycated HDL. AIMS We tested whether modification of HDL in vitro by glycolaldehyde (GAD), malondialdehyde (MDA) or glucose affected HDL's anti-inflammatory properties and ability to promote cholesterol efflux. To determine whether similar changes occur in vivo, we examined modifications of apolipoprotein A1 (APOA1) and APOA2 and anti-inflammatory and cholesterol efflux properties of HDL isolated from subjects with type 1 diabetes in poor glycemic control. RESULTS In vitro modification with both GAD and MDA blunted HDL's ability to inhibit palmitate-induced inflammation and cholesterol efflux in adipocytes. Modification of HDL by glucose had little impact on HDL function, like the response using HDL isolated from subjects with diabetes. Mass spectrophotometric analysis revealed that lysine residues in APOA1 and APOA2 of HDL modified by GAD and MDA in vitro differed from those modified by glucose, which resembled that seen with HDL from patients with type1 diabetes. CONCLUSIONS Modification of lysine residues in HDL by GAD and MDA in vitro does not mirror the HDL glycation in vivo in patients with diabetes, but resembles HDL modified in vitro by glucose.
Collapse
Affiliation(s)
- Diego Gomes Kjerulf
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Shari Wang
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Mohamed Omer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Asha Pathak
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Savitha Subramanian
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Chang Yeop Han
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Chongren Tang
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Laura J den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Baohai Shao
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
31
|
Márquez AB, Nazir S, van der Vorst EP. High-Density Lipoprotein Modifications: A Pathological Consequence or Cause of Disease Progression? Biomedicines 2020; 8:biomedicines8120549. [PMID: 33260660 PMCID: PMC7759904 DOI: 10.3390/biomedicines8120549] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
High-density lipoprotein (HDL) is well-known for its cardioprotective effects, as it possesses anti-inflammatory, anti-oxidative, anti-thrombotic, and cytoprotective properties. Traditionally, studies and therapeutic approaches have focused on raising HDL cholesterol levels. Recently, it became evident that, not HDL cholesterol, but HDL composition and functionality, is probably a more fruitful target. In disorders, such as chronic kidney disease or cardiovascular diseases, it has been observed that HDL is modified and becomes dysfunctional. There are different modification that can occur, such as serum amyloid, an enrichment and oxidation, carbamylation, and glycation of key proteins. Additionally, the composition of HDL can be affected by changes to enzymes such as cholesterol ester transfer protein (CETP), lecithin-cholesterol acyltransferase (LCAT), and phospholipid transfer protein (PLTP) or by modification to other important components. This review will highlight some main modifications to HDL and discuss whether these modifications are purely a consequential result of pathology or are actually involved in the pathology itself and have a causal role. Therefore, HDL composition may present a molecular target for the amelioration of certain diseases, but more information is needed to determine to what extent HDL modifications play a causal role in disease development.
Collapse
Affiliation(s)
- Andrea Bonnin Márquez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Sumra Nazir
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P.C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (A.B.M.); (S.N.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, 80336 Munich, Germany
- Correspondence: ; Tel.: +49-241-80-36914
| |
Collapse
|
32
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
33
|
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
|
34
|
Current Understanding of the Relationship of HDL Composition, Structure and Function to Their Cardioprotective Properties in Chronic Kidney Disease. Biomolecules 2020; 10:biom10091348. [PMID: 32967334 PMCID: PMC7564231 DOI: 10.3390/biom10091348] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
In the general population, the ability of high-density lipoproteins (HDLs) to promote cholesterol efflux is a predictor of cardiovascular events, independently of HDL cholesterol levels. Although patients with chronic kidney disease (CKD) have a high burden of cardiovascular morbidity and mortality, neither serum levels of HDL cholesterol, nor cholesterol efflux capacity associate with cardiovascular events. Important for the following discussion on the role of HDL in CKD is the notion that traditional atherosclerotic cardiovascular risk factors only partially account for this increased incidence of cardiovascular disease in CKD. As a potential explanation, across the spectrum of cardiovascular disease, the relative contribution of atherosclerotic cardiovascular disease becomes less important with advanced CKD. Impaired renal function directly affects the metabolism, composition and functionality of HDL particles. HDLs themselves are a heterogeneous population of particles with distinct sizes and protein composition, all of them affecting the functionality of HDL. Therefore, a more specific approach investigating the functional and compositional features of HDL subclasses might be a valuable strategy to decipher the potential link between HDL, cardiovascular disease and CKD. This review summarizes the current understanding of the relationship of HDL composition, metabolism and function to their cardio-protective properties in CKD, with a focus on CKD-induced changes in the HDL proteome and reverse cholesterol transport capacity. We also will highlight the gaps in the current knowledge regarding important aspects of HDL biology.
Collapse
|
35
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
36
|
Abstract
Cardiovascular disease and infections are major causes for the high incidence of morbidity and mortality of patients with chronic kidney disease. Both complications are directly or indirectly associated with disturbed functions or altered apoptotic rates of polymorphonuclear leukocytes, monocytes, lymphocytes, and dendritic cells. Normal responses of immune cells can be reduced, leading to infectious diseases or pre-activated/primed, giving rise to inflammation and subsequently to cardiovascular disease. This review summarizes the impact of kidney dysfunction on the immune system. Renal failure results in disturbed renal metabolic activities with reduced renin, erythropoietin, and vitamin D production, which adversely affects the immune system. Decreased kidney function also leads to reduced glomerular filtration and the retention of uremic toxins. A large number of uremic toxins with detrimental effects on immune cells have been identified. Besides small water-soluble and protein-bound compounds originating from the intestinal microbiome, several molecules in the middle molecular range, e.g., immunoglobulin light chains, retinol-binding protein, the neuropeptides Met-enkephalin and neuropeptide Y, endothelin-1, and the adipokines leptin and resistin, adversely affect immune cells. Posttranslational modifications such as carbamoylation, advanced glycation products, and oxidative modifications contribute to uremic toxicity. Furthermore, high-density lipoprotein from uremic patients has an altered protein profile and thereby loses its anti-inflammatory properties.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
37
|
High-Density Lipoprotein (HDL) Inhibits Serum Amyloid A (SAA)-Induced Vascular and Renal Dysfunctions in Apolipoprotein E-Deficient Mice. Int J Mol Sci 2020; 21:ijms21041316. [PMID: 32075280 PMCID: PMC7072968 DOI: 10.3390/ijms21041316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 01/09/2023] Open
Abstract
Serum amyloid A (SAA) promotes endothelial inflammation and dysfunction that is associated with cardiovascular disease and renal pathologies. SAA is an apoprotein for high-density lipoprotein (HDL) and its sequestration to HDL diminishes SAA bioactivity. Herein we investigated the effect of co-supplementing HDL on SAA-mediated changes to vascular and renal function in apolipoprotein E-deficient (ApoE-/-) mice in the absence of a high-fat diet. Male ApoE-/- mice received recombinant human SAA or vehicle (control) by intraperitoneal (i.p.) injection every three days for two weeks with or without freshly isolated human HDL supplemented by intravenous (i.v.) injection in the two weeks preceding SAA stimulation. Aorta and kidney were harvested 4 or 18 weeks after commencement of treatment. At 4 weeks after commencement of treatment, SAA increased aortic vascular cell adhesion molecule (VCAM)-1 expression and F2-isoprostane level and decreased cyclic guanosine monophosphate (cGMP), consistent with SAA stimulating endothelial dysfunction and promoting atherosclerosis. SAA also stimulated renal injury and inflammation that manifested as increased urinary protein, kidney injury molecule (KIM)-1, and renal tissue cytokine/chemokine levels as well as increased protein tyrosine chlorination and P38 MAPkinase activation and decreased in Bowman's space, confirming that SAA elicited a pro-inflammatory phenotype in the kidney. At 18 weeks, vascular lesions increased significantly in the cohort of ApoE-/- mice treated with SAA alone. By contrast, pretreatment of mice with HDL decreased SAA pro-inflammatory activity, inhibited SAA enhancement of aortic lesion size and renal function, and prevented changes to glomerular Bowman's space. Taken together, these data indicate that supplemented HDL reduces SAA-mediated endothelial and renal dysfunction in an atherosclerosis-prone mouse model.
Collapse
|
38
|
The Role and Function of HDL in Patients with Chronic Kidney Disease and the Risk of Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21020601. [PMID: 31963445 PMCID: PMC7014265 DOI: 10.3390/ijms21020601] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem with steadily increasing occurrence. Significantly elevated cardiovascular morbidity and mortality have been observed in CKD. Cardiovascular diseases are the most important and frequent cause of death of CKD patients globally. The presence of CKD is related to disturbances in lipoprotein metabolism whose consequences are dyslipidemia and the accumulation of atherogenic particles. CKD not only fuels the reduction of high-density lipoprotein (HDL) cholesterol concentration, but also it modifies the composition of this lipoprotein. The key role of HDL is the participation in reverse cholesterol transport from peripheral tissues to the liver. Moreover, HDL prevents the oxidation of low-density lipoprotein (LDL) cholesterol by reactive oxygen species (ROS) and protects against the adverse effects of oxidized LDL (ox-LDL) on the endothelium. Numerous studies have demonstrated the ability of HDL to promote the production of nitric oxide (NO) by endothelial cells (ECs) and to exert antiapoptotic and anti-inflammatory effects. Increasing evidence suggests that in patients with chronic inflammatory disorders, HDLs may lose important antiatherosclerotic properties and become dysfunctional. So far, no therapeutic strategy to raise HDL, or alter the ratio of HDL subfractions, has been successful in slowing the progression of CKD or reducing cardiovascular disease in patients either with or without CKD.
Collapse
|
39
|
Shi R, Wu B, Niu Z, Sun H, Hu F. Nomogram Based on Risk Factors for Type 2 Diabetes Mellitus Patients with Coronary Heart Disease. Diabetes Metab Syndr Obes 2020; 13:5025-5036. [PMID: 33376372 PMCID: PMC7756175 DOI: 10.2147/dmso.s273880] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/21/2020] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION This study aimed to study risk factors for coronary heart disease (CHD) in type 2 diabetes mellitus (T2DM) patients and establish a clinical prediction model. RESEARCH DESIGN AND METHODS A total of 3402 T2DM patients were diagnosed by clinical doctors and recorded in the electronic medical record system (EMRS) of six Community Health Center Hospitals from 2015 to 2017, including the communities of Huamu, Jinyang, Yinhang, Siping, Sanlin and Daqiao. From September 2018 to September 2019, 3361 patients (41 patients were missing) were investigated using a questionnaire, physical examination, and biochemical index test. After excluding the uncompleted data, 3214 participants were included in the study and randomly divided into a training set (n = 2252) and a validation set (n = 962) at a ratio of 3:1. Through lead absolute shrinkage and selection operator (LASSO) regression analysis and logistic regression analysis of the training set, risk factors were determined and included in a nomogram. The C-index, receiver operating characteristic (ROC) curve, calibration plot and decision curve analysis (DCA) were used to validate the distinction, calibration and clinical practicality of the model. RESULTS Age, T2DM duration, hypertension (HTN), hyperuricaemia (HUA), body mass index (BMI), glycosylated haemoglobin A1c (HbA1c), high-density lipoprotein (HDL-C) and low-density lipoprotein (LDL-C) were significant factors in this study. The C-index was 0.750 (0.724-0.776) based on the training set and 0.767 (0.726-0.808) based on the validation set. Through ROC analysis, the set area was 0.750 for the training set and 0.755 for the validation set. The calibration test indicated that the S:P of the prediction model was 0.982 in the training set and 0.499 in the validation set. The decision curve analysis showed that the threshold probability of the model was 16-69% in the training set and 16-73% in the validation set. CONCLUSION Based on community surveys and data analysis, a prediction model of CHD in T2DM patients was established.
Collapse
Affiliation(s)
- Rong Shi
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Birong Wu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zheyun Niu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hui Sun
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Fan Hu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Correspondence: Fan HuSchool of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of ChinaTel/Fax +862151322466 Email
| |
Collapse
|
40
|
Tao L, ShiChuan W, DeTai Z, Lihua H. Evaluation of lipoprotein-associated phospholipase A2, serum amyloid A, and fibrinogen as diagnostic biomarkers for patients with acute cerebral infarction. J Clin Lab Anal 2019; 34:e23084. [PMID: 31713292 PMCID: PMC7083405 DOI: 10.1002/jcla.23084] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022] Open
Abstract
Objective The aim of this study was to explore the clinical values of combined detection of lipoprotein‐associated phospholipase A2 (Lp‐PLA2), serum amyloid A (SAA), and plasma fibrinogen (FIB) in the diagnosis of acute cerebral infarction (ACI). Methods A case‐control study including 100 hospitalized patients with ACI and 47 healthy controls was carried out. The level of Lp‐PLA2, SAA, and FIB was detected, respectively, and their clinical values were analyzed. Carotid lesions and neurological impairment were also analyzed in each patient. Results The level of Lp‐PLA2, SAA, and FIB in the ACI group was significantly higher than that of the controls, and the three biomarkers showed a significant positive correlation and were considered as risk factors for ACI. The area under the curve (AUC) for Lp‐PLA2, SAA, and FIB was 0.858, 0.743, and 0.672, respectively. When three biomarkers were used in combination, the AUC was 0.879. Compared with the other groups, the levels of three biomarkers in bilateral carotid plaque ACI group were all significantly higher. In addition, the level of Lp‐PLA2 and SAA in ACI patients with severe neurological impairment was also significantly higher than that of the mild‐to‐moderate group. Conclusion Lp‐PLA2 combined with SAA and FIB had a high clinical value for rapid diagnosis and prediction of ACI. These biomarkers were also significantly associated with the formation of bilateral carotid atherosclerotic plaques and the severe neurological impairment in ACI patients.
Collapse
Affiliation(s)
- Liang Tao
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang ShiChuan
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang DeTai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Lihua
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Kidney as modulator and target of "good/bad" HDL. Pediatr Nephrol 2019; 34:1683-1695. [PMID: 30291429 PMCID: PMC6450786 DOI: 10.1007/s00467-018-4104-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 10/28/2022]
Abstract
The strong inverse relationship between low levels of high-density lipoproteins (HDLs) and atherosclerotic cardiovascular disease (CVD) led to the designation of HDL as the "good" cholesterol. The atheroprotection is thought to reflect HDL's capacity to efflux cholesterol from macrophages, followed by interaction with other lipoproteins in the plasma, processing by the liver and excretion into bile. However, pharmacologic increases in HDL-C levels have not led to expected clinical benefits, giving rise to the concept of dysfunctional HDL, in which increases in serum HDL-C are not beneficial due to lost or altered HDL functions and transition to "bad" HDL. It is now understood that the cholesterol in HDL, measured by HDL-C, is neither a marker nor the mediator of HDL function, including cholesterol efflux capacity. It is also understood that besides cholesterol efflux, HDL functionality encompasses many other potentially beneficial functions, including antioxidant, anti-inflammatory, antithrombotic, anti-apoptotic, and vascular protective effects that may be critical protective pathways for various cells, including those in the kidney parenchyma. This review highlights advances in our understanding of the role kidneys play in HDL metabolism, including the effects on levels, composition, and functionality of HDL particles, particularly the main HDL protein, apolipoprotein AI (apoAI). We suggest that normal apoAI/HDL in the glomerular filtrate provides beneficial effects, including lymphangiogenesis, that promote resorption of renal interstitial fluid and biological particles. In contrast, dysfunctional apoAI/HDL activates detrimental pathways in tubular epithelial cells and lymphatics that lead to interstitial accumulation of fluid and harmful particles that promote progressive kidney damage.
Collapse
|
42
|
Azabdaftari A, van der Giet M, Schuchardt M, Hennermann JB, Plöckinger U, Querfeld U. The cardiovascular phenotype of adult patients with phenylketonuria. Orphanet J Rare Dis 2019; 14:213. [PMID: 31492166 PMCID: PMC6731621 DOI: 10.1186/s13023-019-1188-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/30/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Patients with Phenylketonuria (PKU) are exposed to multiple cardiovascular risk factors, but the clinical significance of these abnormalities is yet unknown. The purpose of this study was to characterize the cardiovascular phenotype in adult patients with PKU by clinical and dietary data, measurements of biochemical markers, and non-invasive examination of vascular functions. RESULTS Twenty-three adult patients with PKU (age: 18-47 y; 30.8 ± 8.4 y) and 28 healthy controls (age: 18-47 y; 30.1 ± 9.1 y) were included in this study. PKU patients had significantly higher systolic and diastolic blood pressure, increased resting heart rate and a higher body mass index. Total cholesterol and non-HDL cholesterol levels were significantly increased in PKU patients, whereas plasma levels of HDL cholesterol and its subfraction HDL2 (but not HDL3) were significantly decreased. The inflammatory markers C-reactive protein and serum amyloid A protein and the serum oxidative stress marker malondialdehyde were significantly higher in patients with PKU. Venous occlusion plethysmography showed marked reduction in post-ischemic blood flow and the carotid to femoral pulse wave velocity was significantly increased demonstrating endothelial dysfunction and increased vascular stiffness. CONCLUSIONS This study shows that the cardiovascular phenotype of adult PKU patients is characterized by an accumulation of traditional cardiovascular risk factors, high levels of inflammatory and oxidative stress markers, endothelial dysfunction and vascular stiffness. These data indicate the need for early cardiovascular risk reduction in patients with PKU.
Collapse
Affiliation(s)
- Aline Azabdaftari
- Department of Pediatrics, Division of Gastroenterology, Nephrology and Metabolic Diseases, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augstenburger Platz 1, 13353, Berlin, Germany
| | - Markus van der Giet
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Julia B Hennermann
- Villa Metabolica, Department of Pediatric and Adolescent Medicine, University Medical Center Mainz, Langenbeckstr, 1, 55131, Mainz, Germany
| | - Ursula Plöckinger
- Interdisciplinary Center of Metabolism: Endocrinology, Diabetes and Metabolism, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Uwe Querfeld
- Department of Pediatrics, Division of Gastroenterology, Nephrology and Metabolic Diseases, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augstenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
43
|
Leung ET, Raboin MJ, McKelvey J, Graham A, Lewis A, Prongay K, Cohen AM, Vinson A. Modelling disease risk for amyloid A (AA) amyloidosis in non-human primates using machine learning. Amyloid 2019; 26:139-147. [PMID: 31210531 PMCID: PMC6667354 DOI: 10.1080/13506129.2019.1625038] [Citation(s) in RCA: 4] [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: 07/11/2018] [Revised: 05/12/2019] [Accepted: 05/26/2019] [Indexed: 02/02/2023]
Abstract
Objective: Amyloid A (AA) amyloidosis is found in humans and non-human primates, but quantifying disease risk prior to clinical symptoms is challenging. We applied machine learning to identify the best predictors of amyloidosis in rhesus macaques from available clinical and pathology records. To explore potential biomarkers, we also assessed whether changes in circulating serum amyloid A (SAA) or lipoprotein profiles accompany the disease. Methods: We conducted a retrospective study using 86 cases and 163 controls matched for age and sex. We performed data reduction on 62 clinical, pathological and demographic variables, and applied multivariate modelling and model selection with cross-validation. To test the performance of our final model, we applied it to a replication cohort of 2,775 macaques. Results: The strongest predictors of disease were colitis, gastrointestinal adenocarcinoma, endometriosis, arthritis, trauma, diarrhoea and number of pregnancies. Sensitivity and specificity of the risk model were predicted to be 82%, and were assessed at 79 and 72%, respectively. Total, low density lipoprotein and high density lipoprotein cholesterol levels were significantly lower, and SAA levels and triglyceride-to-HDL ratios were significantly higher in cases versus controls. Conclusion: Machine learning is a powerful approach to identifying macaques at risk of AA amyloidosis, which is accompanied by increased circulating SAA and altered lipoprotein profiles.
Collapse
Affiliation(s)
- Eric T. Leung
- Div. of Bioinformatics and Computational Biology, Dept. of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon
| | - Michael J. Raboin
- Primate Genetics Section, Oregon National Primate Research Center, Beaverton, Oregon
| | - Jessica McKelvey
- Div. of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Adam Graham
- Div. of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Anne Lewis
- Div. of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Kamm Prongay
- Div. of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Aaron M. Cohen
- Div. of Bioinformatics and Computational Biology, Dept. of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon
| | - Amanda Vinson
- Div. of Bioinformatics and Computational Biology, Dept. of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon
- Primate Genetics Section, Oregon National Primate Research Center, Beaverton, Oregon
- Div. of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Acute phase serum amyloid A (SAA) is persistently elevated in chronic inflammatory conditions, and elevated levels predict cardiovascular risk in humans. More recently, murine studies have demonstrated that over-expression of SAA increases and deficiency/suppression of SAA attenuates atherosclerosis. Thus, beyond being a biomarker, SAA appears to play a causal role in atherogenesis. The purpose of this review is to summarize the data supporting SAA as a key player in atherosclerosis development. RECENT FINDINGS A number of pro-inflammatory and pro-atherogenic activities have been ascribed to SAA. However, the literature is conflicted, as recombinant SAA, and/or lipid-free SAA, used in many of the earlier studies, do not reflect the activity of native human or murine SAA, which exists largely lipid-associated. Recent literatures demonstrate that SAA activates the NLRP3 inflammasome, alters vascular function, affects HDL function, and increases thrombosis. Importantly, SAA activity appears to be regulated by its lipid association, and HDL may serve to sequester and limit SAA activity. SUMMARY SAA has many pro-inflammatory and pro-atherogenic activities, is clearly demonstrated to affect atherosclerosis development, and may be a candidate target for clinical trials in cardiovascular diseases.
Collapse
Affiliation(s)
- Preetha Shridas
- Department of Internal Medicine
- Saha Cardiovascular Research Center
- Barnstable Brown Diabetes Center and University of Kentucky
| | - Lisa R Tannock
- Department of Internal Medicine
- Saha Cardiovascular Research Center
- Barnstable Brown Diabetes Center and University of Kentucky
- Veterans Affairs Lexington, Lexington, Kentucky, USA
| |
Collapse
|
45
|
Takase H, Tanaka M, Nakamura Y, Morita SY, Yamada T, Mukai T. Effects of lipid composition on the structural properties of human serum amyloid A in reconstituted high-density lipoprotein particles. Chem Phys Lipids 2019; 221:8-14. [DOI: 10.1016/j.chemphyslip.2019.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022]
|
46
|
Hung AM, Tsuchida Y, Nowak KL, Sarkar S, Chonchol M, Whitfield V, Salas N, Dikalova A, Yancey PG, Huang J, Linton MF, Ikizler TA, Kon V. IL-1 Inhibition and Function of the HDL-Containing Fraction of Plasma in Patients with Stages 3 to 5 CKD. Clin J Am Soc Nephrol 2019; 14:702-711. [PMID: 31015261 PMCID: PMC6500942 DOI: 10.2215/cjn.04360418] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 03/15/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVES Systemic inflammation modulates cardiovascular disease risk and functionality of HDL in the setting of CKD. Whether interventions that modify systemic inflammation can improve HDL function in CKD is unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We conducted a post hoc analysis of two randomized, clinical trials, IL-1 trap in participants with GFR 15-59 ml/min per 1.73 m2 (study A) and IL-1 receptor antagonist in participants on maintenance hemodialysis (study B), to evaluate if IL-1 blockade had improved the anti-inflammatory activity (IL-6, TNF-α, and Nod-like receptor protein 3), antioxidant function (superoxide production), and net cholesterol efflux capacity of HDL. HDL function was measured using LPS-stimulated THP-1 macrophages or peritoneal macrophages of apoE-deficient mice exposed to the apoB-depleted, HDL-containing fraction obtained from the plasma of the study participants, collected before and after the interventions to block IL-1 effects. Analysis of covariance was used for between group comparisons. RESULTS The mean age of the participants was 60±13 years, 72% (n=33) were men, and 39% (n=18) were black. There were 32 CKD (16 IL-1 trap and 16 placebo) and 14 maintenance hemodialysis (7 IL-1 receptor antagonist and 7 placebo) participants. Compared with placebo, IL-1 inhibition, in study A and B reduced cellular expression of TNF-α by 15% (P=0.05) and 64% (P=0.02), IL-6 by 38% (P=0.004) and 56% (P=0.08), and Nod-like receptor protein 3 by 16% (P=0.01) and 25% (P=0.02), respectively. The intervention blunted superoxide production in the treated arm compared with placebo, with the values being higher by 17% in the placebo arm in study A (P<0.001) and 12% in the placebo arm in study B (P=0.004). Net cholesterol efflux capacity was not affected by either intervention. CONCLUSIONS IL-1 blockade improves the anti-inflammatory and antioxidative properties of the HDL-containing fraction of plasma in patients with stages 3-5 CKD, including those on maintenance hemodialysis.
Collapse
Affiliation(s)
- Adriana M Hung
- Division of Nephrology, Tennessee Valley Healthcare System, Nashville, Tennessee; .,Department of Medicine and
| | - Yohei Tsuchida
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Kristen L Nowak
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | | | - Michel Chonchol
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | | | | | | | | | | | | | - T Alp Ikizler
- Division of Nephrology, Tennessee Valley Healthcare System, Nashville, Tennessee.,Department of Medicine and
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; and
| |
Collapse
|
47
|
Fournier M, Bonneil E, Garofalo C, Grimard G, Laverdière C, Krajinovic M, Drouin S, Sinnett D, Marcil V, Levy E. Altered proteome of high-density lipoproteins from paediatric acute lymphoblastic leukemia survivors. Sci Rep 2019; 9:4268. [PMID: 30862935 PMCID: PMC6414624 DOI: 10.1038/s41598-019-40906-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 01/30/2019] [Indexed: 01/16/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most frequent malignancy in children. With the use of more modern, efficient treatments, 5-year survival has reached more than 90% in this population. However, this achievement comes with many secondary and long-term effects since more than 65% of the survivors experience at least one severe complication, including the metabolic syndrome and cardiovascular diseases. The main objective of the present work was to characterize the composition of HDL particles isolated from pediatric ALL survivors. HDLs from 8 metabolically healthy ALL survivors, 8 metabolically unhealthy ALL survivors and 8 age- and gender-matched controls were analyzed. The HDL fraction from the survivors contained less cholesterol than the controls. In addition, proteomic analyses revealed an enrichment of pro-thrombotic (e.g., fibrinogen) and pro-inflammatory (e.g., amyloid A) proteins in the HDLs deriving from metabolically unhealthy survivors. These results indicate an alteration in the composition of lipid and protein content of HDL from childhood ALL survivors with metabolic disorders. Although more work is needed to validate the functionality of these HDLs, the data seem relevant for survivor health given the detection of potential biomarkers related to HDL metabolism and functionality in cancer.
Collapse
Affiliation(s)
- Maryse Fournier
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Eric Bonneil
- Institute of Research in Immunology and Cancer, Université de Montréal, QC, H3C 3J7, Montréal, Canada
| | - Carole Garofalo
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Guy Grimard
- Department of Pediatrics, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Caroline Laverdière
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada.,Department of Pediatrics, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Maja Krajinovic
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada.,Department of Pediatrics, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Simon Drouin
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Daniel Sinnett
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada.,Department of Pediatrics, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Valérie Marcil
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada
| | - Emile Levy
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada. .,Department of Nutrition, Université de Montréal, Montreal, H3T 1C5, Quebec, Canada.
| |
Collapse
|
48
|
Schuchardt M, Prüfer N, Tu Y, Herrmann J, Hu XP, Chebli S, Dahlke K, Zidek W, van der Giet M, Tölle M. Dysfunctional high-density lipoprotein activates toll-like receptors via serum amyloid A in vascular smooth muscle cells. Sci Rep 2019; 9:3421. [PMID: 30833653 PMCID: PMC6399289 DOI: 10.1038/s41598-019-39846-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 02/01/2019] [Indexed: 01/12/2023] Open
Abstract
Serum amyloid A (SAA) is an uremic toxin and acute phase protein. It accumulates under inflammatory conditions associated with high cardiovascular morbidity and mortality in patients with sepsis or end-stage renal disease (ESRD). SAA is an apolipoprotein of the high-density lipoprotein (HDL). SAA accumulation turns HDL from an anti-inflammatory to a pro-inflammatory particle. SAA activates monocyte chemoattractant protein-1 (MCP-1) in vascular smooth muscle cells. However, the SAA receptor-mediated signaling pathway in vascular cells is poorly understood. Therefore, the SAA-mediated signaling pathway for MCP-1 production was investigated in this study. The SAA-induced MCP-1 production is dependent on the activation of TLR2 and TLR4 as determined by studies with specific receptor antagonists and agonists or siRNA approach. Experiments were confirmed in tissues from TLR2 knockout, TLR4 deficient and TLR2 knock-out/TLR4 deficient mice. The intracellular signaling pathway is IκBα and subsequently NFκB dependent. The MCP-1 production induced by SAA-enriched HDL and HDL isolated from septic patients with high SAA content is also TLR2 and TLR4 dependent. Taken together, the TLR2 and TLR4 receptors are functional SAA receptors mediating MCP-1 release. Furthermore, the TLR2 and TLR4 are receptors for dysfunctional HDL. These results give a further inside in SAA as uremic toxin involved in uremia-related pro-inflammatory response in the vascular wall.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Nicole Prüfer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Yuexing Tu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany.,Zhejiang Provincial People´s Hospital, Intensive Care Unit, Hangzhou, China
| | - Jaqueline Herrmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Xiu-Ping Hu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Sarah Chebli
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Katja Dahlke
- Deutsches Institut für Ernaehrungsforschung, Department of Gastrointestinal Microbiology, Arthur-Scheunert-Allee 114-116, 14558, Nuthethal, Germany
| | - Walter Zidek
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Markus van der Giet
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Markus Tölle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| |
Collapse
|
49
|
High-Density Lipoprotein from Chronic Kidney Disease Patients Modulates Polymorphonuclear Leukocytes. Toxins (Basel) 2019; 11:toxins11020073. [PMID: 30717079 PMCID: PMC6409858 DOI: 10.3390/toxins11020073] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 12/24/2022] Open
Abstract
The anti-inflammatory properties of high-density lipoproteins (HDL) are lost in uremia. These HDL may show pro-inflammatory features partially as a result of changed protein composition. Alterations of polymorphonuclear leukocytes (PMNLs) in chronic kidney disease (CKD) may contribute to chronic inflammation and high vascular risk. We investigated if HDL from uremic patients is related to systemic inflammation by interfering with PMNL function. PMNL apoptosis was investigated by assessing morphological features and DNA content. CD11b surface expression was quantified by flow cytometry. Oxidative burst was measured via cytochrome c reduction assay. Chemotaxis was assessed by using an under-agarose migration assay. We found that HDL from CKD and hemodialysis (HD) patients significantly attenuated PMNL apoptosis, whereas HDL isolated from healthy subjects had no effect on PMNL apoptosis. The use of signal transduction inhibitors indicated that uremic HDL exerts anti-apoptotic effects by activating pathways involving phosphoinositide 3-kinase and extracellular-signal regulated kinase. Healthy HDL attenuated the surface expression of CD11b, whereas HDL from CKD and HD patients had no effect. All tested isolates increased the stimulation of oxidative burst, but did not affect PMNL chemotactic movement. In conclusion, HDL may contribute to the systemic inflammation in uremic patients by modulating PMNL functions.
Collapse
|
50
|
The effect of chronic kidney disease on lipid metabolism. Int Urol Nephrol 2018; 51:265-277. [DOI: 10.1007/s11255-018-2047-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/27/2018] [Indexed: 12/26/2022]
|