1
|
Stadler JT, Borenich A, Pammer A, Emrich IE, Habisch H, Madl T, Heine GH, Marsche G. Association of Small HDL Subclasses with Mortality Risk in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:1511. [PMID: 39765838 PMCID: PMC11673888 DOI: 10.3390/antiox13121511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
High-density lipoproteins (HDL) exist in various subclasses, with smaller HDL particles possessing the highest anti-oxidative and anti-inflammatory properties. Understanding the role of these specific subclasses in chronic kidney disease (CKD) could provide valuable insights into disease progression and potential therapeutic targets. In the present study, we assessed HDL subclass composition in 463 patients with CKD stage 2-4 using nuclear magnetic resonance spectroscopy. Over a mean follow-up period of 5.0 years, 18.6% of patients died. Compared to survivors, deceased patients exhibited significantly lower levels of cholesterol, ApoA-I, and ApoA-II within the small and extra-small (XS) HDL subclasses. Multivariable Cox regression analysis, adjusted for traditional cardiovascular and renal risk factors, demonstrated that reduced levels of XS-HDL-cholesterol, XS-HDL-ApoA-I, and XS-HDL-ApoA-II were independently associated with an increased risk of mortality. Furthermore, receiver operating characteristic analysis identified XS-HDL-ApoA-II as the most potent prognostic marker for mortality. In conclusion, reduced small and XS-HDL subclasses, especially XS-HDL-ApoA-II, are strongly associated with increased all-cause mortality risk in CKD patients. Assessment of HDL subclass distribution could provide valuable clinical information and help identify patients at high risk.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (A.P.)
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Beethovenstraße 8, 8010 Graz, Austria
| | - Andrea Borenich
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Auenbruggerplatz 2, 8036 Graz, Austria;
| | - Anja Pammer
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (A.P.)
| | - Insa E. Emrich
- Faculty of Medicine, Saarland University, 66421 Saarbrücken, Germany;
| | - Hansjörg Habisch
- Division of Medical Chemistry, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (H.H.); (T.M.)
| | - Tobias Madl
- Division of Medical Chemistry, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (H.H.); (T.M.)
- BioTechMed Graz, 8010 Graz, Austria
| | - Gunnar H. Heine
- Faculty of Medicine, Saarland University, 66421 Saarbrücken, Germany;
- Department of Nephrology, Agaplesion Markus Krankenhaus, 60431 Frankfurt am Main, Germany
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (A.P.)
- BioTechMed Graz, 8010 Graz, Austria
| |
Collapse
|
2
|
Giacaglia MB, Pires V, Santana MFM, Passarelli M. Unraveling the Pleiotropic Role of High-Density Lipoproteins (HDLs) in Autoimmune Rheumatic Diseases. Int J Rheumatol 2024; 2024:1896817. [PMID: 39574464 PMCID: PMC11581784 DOI: 10.1155/2024/1896817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Autoimmune rheumatic diseases (ARDs) exhibit an elevated incidence of cardiovascular disease (CVD). The elevation of inflammatory and immune stress accompanying ARDs contributes to atherosclerosis development and alterations in lipid metabolism and lipoprotein profile add to cardiovascular (CV) risk. The plasma concentration of high-density lipoprotein cholesterol (HDLc) is inversely related to CVD and serves as a discriminator of CV risk. However, this association is not unequivocal, and changes in HDL functionality appear to emerge as a better indicator of CV risk, albeit difficult to measure and monitor clinically. The modulation of HDLc itself can bring benefits in controlling autoimmunity and reducing ARD activity. Understanding HDL function and each peculiarity involved in ARDs enables to seek means to prevent ischemic outcomes associated with CVD, in the face of the residual CV risk persisting even with controlled disease activity and classic risk factors. By comprehending HDL's structural and functional nuances, it will be possible to develop more effective strategies to manage the evolution and outcomes of ARDs. It is also necessary to standardize diagnostic methods and establish different markers for each specific disease allowing the design of intervention strategies to restore HDL functionality, reduce residual CV, and prevent, alleviate, or even suppress ARD activity.
Collapse
Affiliation(s)
- Marcia B. Giacaglia
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
| | - Vitória Pires
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Monique F. M. Santana
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Marisa Passarelli
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| |
Collapse
|
3
|
Mosalmanzadeh N, Pence BD. Oxidized Low-Density Lipoprotein and Its Role in Immunometabolism. Int J Mol Sci 2024; 25:11386. [PMID: 39518939 PMCID: PMC11545486 DOI: 10.3390/ijms252111386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Modified cholesterols such as oxidized low-density lipoprotein (OxLDL) contribute to atherosclerosis and other disorders through the promotion of foam cell formation and inflammation. In recent years, it has become evident that immune cell responses to inflammatory molecules such as OxLDLs depend on cellular metabolic functions. This review examines the known effects of OxLDL on immunometabolism and immune cell responses in atherosclerosis and several other diseases. We additionally provide context on the relationship between OxLDL and aging/senescence and identify gaps in the literature and our current understanding in these areas.
Collapse
Affiliation(s)
| | - Brandt D. Pence
- College of Health Sciences and Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38111, USA
| |
Collapse
|
4
|
Frostegård J. Lipids in SLE. Rheumatology (Oxford) 2024; 63:2613-2614. [PMID: 38775457 DOI: 10.1093/rheumatology/keae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2024] [Indexed: 10/02/2024] Open
|
5
|
Rani A, Stadler JT, Marsche G. HDL-based therapeutics: A promising frontier in combating viral and bacterial infections. Pharmacol Ther 2024; 260:108684. [PMID: 38964560 DOI: 10.1016/j.pharmthera.2024.108684] [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: 02/26/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Low levels of high-density lipoprotein (HDL) and impaired HDL functionality have been consistently associated with increased susceptibility to infection and its serious consequences. This has been attributed to the critical role of HDL in maintaining cellular lipid homeostasis, which is essential for the proper functioning of immune and structural cells. HDL, a multifunctional particle, exerts pleiotropic effects in host defense against pathogens. It functions as a natural nanoparticle, capable of sequestering and neutralizing potentially harmful substances like bacterial lipopolysaccharides. HDL possesses antiviral activity, preventing viruses from entering or fusing with host cells, thereby halting their replication cycle. Understanding the complex relationship between HDL and the immune system may reveal innovative targets for developing new treatments to combat infectious diseases and improve patient outcomes. This review aims to emphasize the role of HDL in influencing the course of bacterial and viral infections and its and its therapeutic potential.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Styria, Austria.
| |
Collapse
|
6
|
Atehortua L, Sean Davidson W, Chougnet CA. Interactions Between HDL and CD4+ T Cells: A Novel Understanding of HDL Anti-Inflammatory Properties. Arterioscler Thromb Vasc Biol 2024; 44:1191-1201. [PMID: 38660807 PMCID: PMC11111342 DOI: 10.1161/atvbaha.124.320851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Several studies in animal models and human cohorts have recently suggested that HDLs (high-density lipoproteins) not only modulate innate immune responses but also adaptative immune responses, particularly CD4+ T cells. CD4+ T cells are central effectors and regulators of the adaptive immune system, and any alterations in their homeostasis contribute to the pathogenesis of cardiovascular diseases, autoimmunity, and inflammatory diseases. In this review, we focus on how HDLs and their components affect CD4+ T-cell homeostasis by modulating cholesterol efflux, immune synapsis, proliferation, differentiation, oxidative stress, and apoptosis. While the effects of apoB-containing lipoproteins on T cells have been relatively well established, this review focuses specifically on new connections between HDL and CD4+ T cells. We present a model where HDL may modulate T cells through both direct and indirect mechanisms.
Collapse
Affiliation(s)
- Laura Atehortua
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH
| | - W. Sean Davidson
- Division of Experimental Pathology, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
7
|
Cao L, Wu C, Liu M, Zhang W, Chen H, Wang R, He Z. The association between monocyte-to-high-density lipoprotein ratio and hyperuricemia: Results from 2009 to 2018. Medicine (Baltimore) 2024; 103:e37713. [PMID: 38669360 PMCID: PMC11049789 DOI: 10.1097/md.0000000000037713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/04/2024] [Indexed: 04/28/2024] Open
Abstract
Previous research has suggested that the monocyte-to-high-density lipoprotein ratio (MHR), an emerging inflammatory biomarker, holds promise in predicting the prevalence of various cardiovascular and metabolic diseases. However, earlier investigations were constrained by the relatively modest sample sizes. This study endeavored to expand the sample size and conduct a more comprehensive exploration of the potential relationship between MHR and hyperuricemia. This cross-sectional study incorporated data from participants of the 2009 to 2018 National Health and Nutrition Examination Survey (NHANES) with complete and qualifying information. MHR was determined by calculating the ratio between monocyte count and high-density lipoprotein levels. Various statistical methodologies such as weighted multivariate logistic regression, subgroup analysis, smoothed curve fitting, and threshold analysis, have been used to explore the correlation between hyperuricemia and MHR. The study included a cohort of 17,694 participants, of whom 3512 were diagnosed with hyperuricemia. MHR levels were notably higher in the hyperuricemia group than in the normal group, aligning with an elevated body mass index (BMI). A comprehensive multivariate logistic analysis, accounting for all relevant adjustments, revealed a notable positive correlation between MHR and hyperuricemia (P < .001, OR = 1.98, 95% CI: 1.54-2.54). Subgroup analysis indicated that the MHR exhibited an enhanced predictive capacity for identifying hyperuricemia risk, particularly in females (P < .05). Curvilinear and threshold analyses revealed a nonlinear association between MHR and hyperuricemia prevalence, with a notable inflection point at 0.826. In the US population, a clear positive correlation was observed between the MHR and prevalence of hyperuricemia. Importantly, the MHR is a more robust predictor of hyperuricemia risk in females. Further investigations are required to confirm these findings.
Collapse
Affiliation(s)
- Lei Cao
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Chunwei Wu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Miao Liu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wenlong Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hailong Chen
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ruolin Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ze He
- Department of Endocrinology and Metabolism, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
8
|
Yang L, Wang Y, Xu Y, Li K, Yin R, Zhang L, Wang D, Wei L, Lang J, Cheng Y, Wang L, Ke J, Zhao D. ANGPTL3 is a novel HDL component that regulates HDL function. J Transl Med 2024; 22:263. [PMID: 38462608 PMCID: PMC10926621 DOI: 10.1186/s12967-024-05032-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/24/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Angiopoietin-like protein 3 (ANGPTL3) is secreted by hepatocytes and inhibits lipoprotein lipase and endothelial lipase activity. Previous studies reported the correlation between plasma ANGPTL3 levels and high-density lipoprotein (HDL). Recently ANGPTL3 was found to preferentially bind to HDL in healthy human circulation. Here, we examined whether ANGPTL3, as a component of HDL, modulates HDL function and affects HDL other components in human and mice with non-diabetes or type 2 diabetes mellitus. METHODS HDL was isolated from the plasma of female non-diabetic subjects and type-2 diabetic mellitus (T2DM) patients. Immunoprecipitation, western blot, and ELISA assays were used to examine ANGPTL3 levels in HDL. Db/m and db/db mice, AAV virus mediated ANGPTL3 overexpression and knockdown models and ANGPTL3 knockout mice were used. The cholesterol efflux capacity induced by HDL was analyzed in macrophages preloaded with fluorescent cholesterol. The anti-inflammation capacity of HDL was assessed using flow cytometry to measure VCAM-1 and ICAM-1 expression levels in TNF-α-stimulated endothelial cells pretreated with HDL. RESULTS ANGPTL3 was found to bind to HDL and be a component of HDL in both non-diabetic subjects and T2DM patients. Flag-ANGPTL3 was found in the HDL of transgenic mice overexpressing Flag-ANGPTL3. ANGPLT3 of HDL was positively associated with cholesterol efflux in female non-diabetic controls (r = 0.4102, p = 0.0117) but not in female T2DM patients (r = - 0.1725, p = 0.3224). Lower ANGPTL3 levels of HDL were found in diabetic (db/db) mice compared to control (db/m) mice and were associated with reduced cholesterol efflux and inhibition of VCAM-1 and ICAM-1 expression in endothelial cells (p < 0.05 for all). Following AAV-mediated ANGPTL3 cDNA transfer in db/db mice, ANGPTL3 levels were found to be increased in HDL, and corresponded to increased cholesterol efflux and decreased ICAM-1 expression. In contrast, knockdown of ANGPTL3 levels in HDL by AAV-mediated shRNA transfer led to a reduction in HDL function (p < 0.05 for both). Plasma total cholesterol, total triglycerides, HDL-c, protein components of HDL and the cholesterol efflux function of HDL were lower in ANGPTL3-/- mice than ANGPTL3+/+ mice, suggesting that ANGPTL3 in HDL may regulate HDL function by disrupting the balance of protein components in HDL. CONCLUSION ANGPTL3 was identified as a component of HDL in humans and mice. ANGPTL3 of HDL regulated cholesterol efflux and the anti-inflammatory functions of HDL in T2DM mice. Both the protein components of HDL and cholesterol efflux capacity of HDL were decreased in ANGPTL3-/- mice. Our findings suggest that ANGPTL3 in HDL may regulate HDL function by disrupting the balance of protein components in HDL. Our study contributes to a more comprehensive understanding of the role of ANGPTL3 in lipid metabolism.
Collapse
Affiliation(s)
- Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yan Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yongsong Xu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Kun Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Ruili Yin
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Di Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lingling Wei
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jianan Lang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yanan Cheng
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lu Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China.
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China.
| |
Collapse
|
9
|
Alkafaas SS, Elsalahaty MI, Ismail DF, Radwan MA, Elkafas SS, Loutfy SA, Elshazli RM, Baazaoui N, Ahmed AE, Hafez W, Diab M, Sakran M, El-Saadony MT, El-Tarabily KA, Kamal HK, Hessien M. The emerging roles of sphingosine 1-phosphate and SphK1 in cancer resistance: a promising therapeutic target. Cancer Cell Int 2024; 24:89. [PMID: 38419070 PMCID: PMC10903003 DOI: 10.1186/s12935-024-03221-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Cancer chemoresistance is a problematic dilemma that significantly restrains numerous cancer management protocols. It can promote cancer recurrence, spreading of cancer, and finally, mortality. Accordingly, enhancing the responsiveness of cancer cells towards chemotherapies could be a vital approach to overcoming cancer chemoresistance. Tumour cells express a high level of sphingosine kinase-1 (SphK1), which acts as a protooncogenic factor and is responsible for the synthesis of sphingosine-1 phosphate (S1P). S1P is released through a Human ATP-binding cassette (ABC) transporter to interact with other phosphosphingolipids components in the interstitial fluid in the tumor microenvironment (TME), provoking communication, progression, invasion, and tumor metastasis. Also, S1P is associated with several impacts, including anti-apoptotic behavior, metastasis, mesenchymal transition (EMT), angiogenesis, and chemotherapy resistance. Recent reports addressed high levels of S1P in several carcinomas, including ovarian, prostate, colorectal, breast, and HCC. Therefore, targeting the S1P/SphK signaling pathway is an emerging therapeutic approach to efficiently attenuate chemoresistance. In this review, we comprehensively discussed S1P functions, metabolism, transport, and signaling. Also, through a bioinformatic framework, we pointed out the alterations of SphK1 gene expression within different cancers with their impact on patient survival, and we demonstrated the protein-protein network of SphK1, elaborating its sparse roles. Furthermore, we made emphasis on different machineries of cancer resistance and the tight link with S1P. We evaluated all publicly available SphK1 inhibitors and their inhibition activity using molecular docking and how SphK1 inhibitors reduce the production of S1P and might reduce chemoresistance, an approach that might be vital in the course of cancer treatment and prognosis.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed I Elsalahaty
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doha F Ismail
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mustafa Ali Radwan
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University-Egypt, New Damietta, 34517, Egypt
| | - Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16th Street, 35233, Khalifa, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo 11511, Egypt
| | - Mohanad Diab
- Burjeel Hospital Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mohamed Sakran
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
10
|
Miroshnichenko S, Pykhtina M, Kotliarova A, Chepurnov A, Beklemishev A. Engineering a New IFN-ApoA-I Fusion Protein with Low Toxicity and Prolonged Action. Molecules 2023; 28:8014. [PMID: 38138504 PMCID: PMC10745500 DOI: 10.3390/molecules28248014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Recombinant human interferon alpha-2b (rIFN) is widely used in antiviral and anticancer immunotherapy. However, the high efficiency of interferon therapy is accompanied by a number of side effects; this problem requires the design of a new class of interferon molecules with reduced cytotoxicity. In this work, IFN was modified via genetic engineering methods by merging it with the blood plasma protein apolipoprotein A-I in order to reduce acute toxicity and improve the pharmacokinetics of IFN. The chimeric protein was obtained via biosynthesis in the yeast P. pastoris. The yield of ryIFN-ApoA-I protein when cultivated on a shaker in flasks was 30 mg/L; protein purification was carried out using reverse-phase chromatography to a purity of 95-97%. The chimeric protein demonstrated complete preservation of the biological activity of IFN in the model of vesicular stomatitis virus and SARS-CoV-2. In addition, the chimeric form had reduced cytotoxicity towards Vero cells and increased cell viability under viral load conditions compared with commercial IFN-a2b preparations. Analysis of the pharmacokinetic profile of ryIFN-ApoA-I after a single subcutaneous injection in mice showed a 1.8-fold increased half-life of the chimeric protein compared with ryIFN.
Collapse
Affiliation(s)
- Svetlana Miroshnichenko
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| | - Mariya Pykhtina
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| | - Anastasiia Kotliarova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia;
| | - Alexander Chepurnov
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| | - Anatoly Beklemishev
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| |
Collapse
|
11
|
Yu M, Dorsey KH, Halseth T, Schwendeman A. Enhancement of Anti-inflammatory Effects of Synthetic High-Density Lipoproteins by Incorporation of Anionic Lipids. Mol Pharm 2023; 20:5454-5462. [PMID: 37781907 PMCID: PMC10916337 DOI: 10.1021/acs.molpharmaceut.3c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Phosphatidylserine (PS) is an anionic phospholipid component in endogenous high-density lipoprotein (HDL). With the intrinsic anti-inflammatory effects of PS and the correlation between PS content and HDL functions, it was hypothesized that incorporating PS would enhance the therapeutic effects of HDL mimetic particles. To test this hypothesis, a series of synthetic high-density lipoproteins (sHDLs) were prepared with an apolipoprotein A-I (ApoA-1) mimetic peptide, 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC), and 1-palmitoyl-2-oleoyl-glycero-3-phospho-l-serine (POPS). Incorporating PS was found to improve the particle stability of sHDLs. Moreover, increasing the PS content in sHDLs enhanced the anti-inflammatory effects on lipopolysaccharide-activated macrophages and endothelial cells. The incorporation of PS had no negative impact on cholesterol efflux capacity, in vivo cholesterol mobilization, and did not affect the pharmacokinetic profiles of sHDLs. Such results suggest the therapeutic potential of PS-containing sHDLs for inflammation resolution in atherosclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kristen Hong Dorsey
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Troy Halseth
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
12
|
Feng J, Huang Y, Huang M, Li X, Amoah K, Huang Y, Jian J. Apolipoprotein Eb (On-ApoEb) protects Oreochromis niloticus against Streptococcus agalactiae infection. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109069. [PMID: 37696347 DOI: 10.1016/j.fsi.2023.109069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Apolipoprotein E (ApoE), a critical targeting protein, has been found to play an essential role in the protection against infection and inflammation. However, the immune functions of ApoE against bacterial infection in fish have not been investigated. In this study, a full-length cDNA for ApoE, named On-ApoEb was cloned from Oreochromis niloticus. The predicted cDNA sequence was 831bp in length and coded for a protein of 276 amino acid residues, which shared 63.87%-98.55% identity with ApoEb from other fishes, and about 22% identity with ApoEb from mammals. On-ApoEb from O. niloticus was highly expressed in the liver and could be activated in the tissues (liver, spleen, brain, and intestine) after infection with Streptococcus agalactiae. Moreover, the results revealed that On-ApoEb could decrease the expression levels of pro-inflammatory factors, immune-related pathways, and apoptosis, while increasing the expression levels of anti-inflammatory factors. Furthermore, On-ApoEb was noted to improve the survival rate and reduce the bacterial load in the liver and spleen. These results suggested that On-ApoEb was connected with immune response and had anti-inflammation and anti-apoptosis activities.
Collapse
Affiliation(s)
- Jiamin Feng
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Yongxiong Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Meiling Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Xing Li
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Kwaku Amoah
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Yu Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| |
Collapse
|
13
|
Jenkins AJ, Carroll LM, Huang MLH, Wen-Loh Y, Mangani A, O'Neal DN, Januszewski AS. Mitochondrial DNA copy number in adults with and without Type 1 diabetes. Diabetes Res Clin Pract 2023; 203:110877. [PMID: 37579994 DOI: 10.1016/j.diabres.2023.110877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
AIMS Mitochondrial damage is implicated in diabetes pathogenesis and complications. Mitochondrial DNA copy number (mtDNA-cn) in human Type 1 diabetes (T1D) studies are lacking. We related mtDNA-cn in T1D and non-diabetic adults (CON) with diabetes complications and risk factors. METHODS Cross-sectional study: 178 T1D, 132 non-diabetic controls. Associations of whole blood mtDNA-cn (qPCR) with complications, inflammation, and C-peptide. RESULTS mtDNA-cn (median (LQ, UQ)) was lower in: T1D vs. CON (271 (189, 348) vs. 320 (264, 410); p < 0.0001); T1D with vs. without kidney disease (238 (180, 309) vs. 294 (198, 364); p = 0.02); and insulin injection vs. pump-users (251 (180, 340) vs. 322 (263, 406); p = 0.008). Significant univariate correlates of mtDNA-cn: T1D: (positive) HDL-C; (negative) fasting glucose, white cell count (WCC), sVCAM-1, sICAM-1; CON: (negative) WHR (waist-hip-ratio). Detectable C-peptide in T1D increased with lowest-highest mtDNA-cn tertiles (54%, 69%, 79%, p = 0.02). Independent determinants of mtDNA-cn: T1D: (positive) HDL-C; (negative) age, sICAM-1; AUROC 0.71; CON: WCC (negative), never smoking, (positive) female, pulse pressure; AUROC 0.74. CONCLUSIONS mtDNA-cn is lower in T1D vs. CON, and in T1D kidney disease. In T1D, mtDNA-cn correlates inversely with age and inflammation, and positively with HDL-C, detectable C-peptide and pump use. Further clinical and basic science studies are merited.
Collapse
Affiliation(s)
- Alicia J Jenkins
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia; Department of Medicine, St. Vincent's Hospital, The University of Melbourne, VIC, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Luke M Carroll
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia
| | - Michael L H Huang
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Yik Wen-Loh
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia
| | - Abubakar Mangani
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia
| | - David N O'Neal
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia; Department of Medicine, St. Vincent's Hospital, The University of Melbourne, VIC, Australia
| | - Andrzej S Januszewski
- NHMRC Clinical Trials Centre, The University of Sydney, NSW, Australia; Department of Medicine, St. Vincent's Hospital, The University of Melbourne, VIC, Australia; Sydney Pharmacy School, The University of Sydney, NSW, Australia.
| |
Collapse
|
14
|
Atehortua L, Morris J, Street SE, Bedel N, Davidson WS, Chougnet CA. Apolipoprotein E-containing HDL decreases caspase-dependent apoptosis of memory regulatory T lymphocytes. J Lipid Res 2023; 64:100425. [PMID: 37579971 PMCID: PMC10507648 DOI: 10.1016/j.jlr.2023.100425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/21/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
Plasma levels of HDL cholesterol are inversely associated with CVD progression. It is becoming increasingly clear that HDL plays important roles in immunity that go beyond its traditionally understood roles in lipid transport. We previously reported that HDL interaction with regulatory T cells (Treg) protected them from apoptosis, which could be a mechanism underlying the broad anti-inflammatory effect of HDL. Herein, we extend our work to show that HDL interacts mainly with memory Treg, particularly with the highly suppressive effector memory Treg, by limiting caspase-dependent apoptosis in an Akt-dependent manner. Reconstitution experiments identified the protein component of HDL as the primary driver of the effect, though the most abundant HDL protein, apolipoprotein A-I (APOA1), was inactive. In contrast, APOE-depleted HDL failed to rescue effector memory Treg, suggesting the critical role of APOE proteins. HDL particles reconstituted with APOE, and synthetic phospholipids blunted Treg apoptosis at physiological concentrations. The APOE3 and APOE4 isoforms were the most efficient. Similar results were obtained when lipid-free recombinant APOEs were tested. Binding experiments showed that lipid-free APOE3 bound to memory Treg but not to naive Treg. Overall, our results show that APOE interaction with Treg results in blunted caspase-dependent apoptosis and increased survival. As dysregulation of HDL-APOE levels has been reported in CVD and obesity, our data bring new insight on how this defect may contribute to these diseases.
Collapse
Affiliation(s)
- Laura Atehortua
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jamie Morris
- Division of Experimental Pathology, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Scott E Street
- Division of Experimental Pathology, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Nicholas Bedel
- Division of Experimental Pathology, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - W Sean Davidson
- Division of Experimental Pathology, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
15
|
Huang Y, Feng J, Li Q, Zhang Z, Jiang B, Amoah K, Huang Y, Jian J. Apolipoprotein A-I (ApoA-I) protects Nile tilapia (Oreochromis niloticus) against bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2023:108925. [PMID: 37414306 DOI: 10.1016/j.fsi.2023.108925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
Apolipoprotein A-I (ApoA-I) is a lipoprotein involved in a variety of physiological and pathological processes. However, the immunomodulatory functions of ApoA-I in fish are not well understood. In this study, ApoA-I from Nile tilapia (Oreochromis niloticus) (On-ApoA-I) was identified, and its function in bacterial infection was investigated. The open reading frame of On-ApoA-I is 792 bp, which codes for a protein containing 263 amino acids. On-ApoA-I shared over 60% sequence similarity with other teleost fish and more than 20% with mammalian ApoA-I. On-ApoA-I was found to be highly expressed in the liver and significantly induced during Streptococcus agalactiae infection by qRT‒PCR analysis. Furthermore, in vivo studies revealed that recombinant On-ApoA-I protein could suppress inflammation and apoptosis and improve the likelihood of surviving bacterial infection. Additionally, On-ApoA-I showed in vitro antimicrobial properties against Gram-positive and Gram-negative bacteria. These findings offer a theoretical basis for further investigations into the role of ApoA-I in fish immunology.
Collapse
Affiliation(s)
- Yongxiong Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Jiamin Feng
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Qi Li
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Zhiqiang Zhang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Baijian Jiang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Kwaku Amoah
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Yu Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| |
Collapse
|
16
|
Abstract
Epidemiologic studies detected an inverse relationship between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major risk factor for ASCVD and suggesting atheroprotective functions of HDL. However, the role of HDL-C as a mediator of risk for ASCVD has been called into question by the failure of HDL-C-raising drugs to reduce cardiovascular events in clinical trials. Progress in understanding the heterogeneous nature of HDL particles in terms of their protein, lipid, and small RNA composition has contributed to the realization that HDL-C levels do not necessarily reflect HDL function. The most examined atheroprotective function of HDL is reverse cholesterol transport, whereby HDL removes cholesterol from plaque macrophage foam cells and delivers it to the liver for processing and excretion into bile. Indeed, in several studies, HDL has shown inverse associations between HDL cholesterol efflux capacity and ASCVD in humans. Inflammation plays a key role in the pathogenesis of atherosclerosis and vulnerable plaque formation, and a fundamental function of HDL is suppression of inflammatory signaling in macrophages and other cells. Oxidation is also a critical process to ASCVD in promoting atherogenic oxidative modifications of LDL (low-density lipoprotein) and cellular inflammation. HDL and its proteins including apoAI (apolipoprotein AI) and PON1 (paraoxonase 1) prevent cellular oxidative stress and LDL modifications. Importantly, HDL in humans with ASCVD is oxidatively modified rendering HDL dysfunctional and proinflammatory. Modification of HDL with reactive carbonyl species, such as malondialdehyde and isolevuglandins, dramatically impairs the antiatherogenic functions of HDL. Importantly, treatment of murine models of atherosclerosis with scavengers of reactive dicarbonyls improves HDL function and reduces systemic inflammation, atherosclerosis development, and features of plaque instability. Here, we discuss the HDL antiatherogenic functions in relation to oxidative modifications and the potential of reactive dicarbonyl scavengers as a therapeutic approach for ASCVD.
Collapse
Affiliation(s)
- MacRae F. Linton
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Patricia G. Yancey
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Huan Tao
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sean S. Davies
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
17
|
Rani A, Marsche G. A Current Update on the Role of HDL-Based Nanomedicine in Targeting Macrophages in Cardiovascular Disease. Pharmaceutics 2023; 15:1504. [PMID: 37242746 PMCID: PMC10221824 DOI: 10.3390/pharmaceutics15051504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
High-density lipoproteins (HDL) are complex endogenous nanoparticles involved in important functions such as reverse cholesterol transport and immunomodulatory activities, ensuring metabolic homeostasis and vascular health. The ability of HDL to interact with a plethora of immune cells and structural cells places it in the center of numerous disease pathophysiologies. However, inflammatory dysregulation can lead to pathogenic remodeling and post-translational modification of HDL, rendering HDL dysfunctional or even pro-inflammatory. Monocytes and macrophages play a critical role in mediating vascular inflammation, such as in coronary artery disease (CAD). The fact that HDL nanoparticles have potent anti-inflammatory effects on mononuclear phagocytes has opened new avenues for the development of nanotherapeutics to restore vascular integrity. HDL infusion therapies are being developed to improve the physiological functions of HDL and to quantitatively restore or increase the native HDL pool. The components and design of HDL-based nanoparticles have evolved significantly since their initial introduction with highly anticipated results in an ongoing phase III clinical trial in subjects with acute coronary syndrome. The understanding of mechanisms involved in HDL-based synthetic nanotherapeutics is critical to their design, therapeutic potential and effectiveness. In this review, we provide a current update on HDL-ApoA-I mimetic nanotherapeutics, highlighting the scope of treating vascular diseases by targeting monocytes and macrophages.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
18
|
Al‐kuraishy HM, Hussien NR, Al‐Niemi MS, Fahad EH, Al‐Buhadily AK, Al‐Gareeb AI, Al‐Hamash SM, Tsagkaris C, Papadakis M, Alexiou A, Batiha GE. SARS-CoV-2 induced HDL dysfunction may affect the host's response to and recovery from COVID-19. Immun Inflamm Dis 2023; 11:e861. [PMID: 37249296 PMCID: PMC10187021 DOI: 10.1002/iid3.861] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
INTRODUCTION Covid-19 is linked with the development of cardio-metabolic disorders, including dyslipidemia, dysregulation of high-density lipoprotein (HDL), and low-density lipoprotein (LDL). Furthermore, SARS-Co-2 infection is associated with noteworthy changes in lipid profile, which is suggested as a possible biomarker to support the diagnosis and management of Covid-19. METHODS This paper adopts the literature review method to obtain information about how Covid-19 affects high-risk group patients and may cause severe and critical effects due to the development of acute lung injury and acute respiratory distress syndrome. A narrative and comprehensive review is presented. RESULTS Reducing HDL in Covid-19 is connected to the disease severity and poor clinical outcomes, suggesting that high HDL serum levels could benefit Covid-19. SARS-CoV-2 binds HDL, and this complex is attached to the co-localized receptors, facilitating viral entry. Therefore, SARS-CoV-2 infection may induce the development of dysfunctional HDL through different mechanisms, including induction of inflammatory and oxidative stress with activation of inflammatory signaling pathways. In turn, the induction of dysfunctional HDL induces the activation of inflammatory signaling pathways and oxidative stress, increasing Covid-19 severity. CONCLUSIONS Covid-19 is linked with the development of cardio-metabolic disorders, including dyslipidemia in general and dysregulation of high-density lipoprotein and low-density lipoprotein. Therefore, the present study aimed to overview the causal relationship between dysfunctional high-density lipoprotein and Covid-19.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Pharmacology, ToxicologyMedicine College of Medicine Al‐Mustansiriyah UniversityBaghdadIraq
| | - Nawar R. Hussien
- Department of Clinical Pharmacy, College of PharmacyAl‐Farahidi UniversityBagdadIraq
| | - Marwa S. Al‐Niemi
- Department of Clinical Pharmacy, College of PharmacyAl‐Farahidi UniversityBagdadIraq
| | | | - Ali K. Al‐Buhadily
- Department of Clinical Pharmacology, Medicine and Therapeutic, Medical Faculty, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Pharmacology, ToxicologyMedicine College of Medicine Al‐Mustansiriyah UniversityBaghdadIraq
| | | | - Christos Tsagkaris
- Department of Health SciencesNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP Med AustriaWienAustria
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
19
|
Cho KH, Nam HS, Kim JE, Na HJ, Del Carmen Dominguez-Horta M, Martinez-Donato G. CIGB-258 Exerts Potent Anti-Inflammatory Activity against Carboxymethyllysine-Induced Acute Inflammation in Hyperlipidemic Zebrafish via the Protection of Apolipoprotein A-I. Int J Mol Sci 2023; 24:ijms24087044. [PMID: 37108210 PMCID: PMC10139093 DOI: 10.3390/ijms24087044] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Inflammation and atherosclerosis are intimately associated via the production of dysfunctional high-density lipoproteins (HDL) and modification of apolipoprotein (apo) A-I. A putative interaction between CIGB-258 and apoA-I was investigated to provide mechanistic insight into the protection of HDL. The protective activity of CIGB-258 was tested in the CML-mediated glycation of apoA-I. The in vivo anti-inflammatory efficacy was compared in paralyzed hyperlipidemic zebrafish and its embryo in the presence of CML. Treatment of CML induced greater glycation extent of HDL/apoA-I and proteolytic degradation of apoA-I. In the presence of CML, however, co-treatment of CIGB-258 inhibited the glycation of apoA-I and protected the degradation of apoA-I, exerting enhanced ferric ion reduction ability. Microinjection of CML (500 ng) into zebrafish embryos resulted in acute death with the lowest survivability with severe developmental defects with interleukin (IL)-6 production. Conversely, a co-injection of CIGB-258 or Tocilizumab produced the highest survivability with a normal development speed and morphology. In hyperlipidemic zebrafish, intraperitoneal injection of CML (500 μg) caused the complete loss of swimming ability and severe acute death with only 13% survivability 3 h post-injection. A co-injection of the CIGB-258 resulted in a 2.2-fold faster recovery of swimming ability than CML alone, with higher survivability of approximately 57%. These results suggest that CIGB-258 protected hyperlipidemic zebrafish from the acute neurotoxicity of CML. Histological analysis showed that the CIGB-258 group had 37% lower infiltration of neutrophils in hepatic tissue and 70% lower fatty liver changes than those of the CML-alone group. The CIGB-258 group exhibited the smallest IL-6 expression in the liver and the lowest blood triglyceride level. CIGB-258 displayed potent anti-inflammatory activity in hyperlipidemic zebrafish by inhibiting apoA-I glycation, promoting rapid recovery from the paralysis of CML toxicity and suppression of IL-6, and lowering fatty liver changes.
Collapse
Affiliation(s)
- Kyung-Hyun Cho
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
- LipoLab, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hyo-Seon Nam
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | - Ji-Eun Kim
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | - Hye-Jee Na
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | | | - Gillian Martinez-Donato
- Center for Genetic Engineering and Biotechnology, Ave 31, e/158 y 190, Playa, La Habana 10600, Cuba
| |
Collapse
|
20
|
Xiong X, Duan Z, Zhou H, Huang G, Niu L, Luo Z, Li W. Correlation of apolipoprotein A-I with T cell subsets and interferon-ү in coronary artery disease. Immun Inflamm Dis 2023; 11:e797. [PMID: 36988256 PMCID: PMC10013138 DOI: 10.1002/iid3.797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND The association of Apolipoprotein A-I (APOAI) with T cell subsets and interferon-ү (IFN-γ) in patients with coronary artery disease (CAD) has been not reported. Thus, this study aimed to investigate the association of APOAI with T cell subsets and IFN-γ in CAD. METHODS This study included a total of 107 patients with CAD including acute coronary syndrome and chronic coronary syndrome. T cell subsets, and CD3-CD56+ natural killer cells were quantified by flow cytometric analysis. The serum concentrations of IFN-ү were measured by enzyme-linked immunosorbent assay. Lipid profiles, C-reactive protein (CRP), and fibrinogen were measured in the clinical laboratory. Clinical data was obtained duration hospitalization. RESULTS The CD4+ T cells were higher in patients of the low-APOAI group ( .05). The high-density lipoprotein cholesterol (HDL-C) was also inversely associated with CD4+ T cells (p < .05), and positively associated with CD8+ T cells (p < .05). Lastly, APOA1 and HDL-C did not correlated with fibrinogen and CRP (p > .05). CONCLUSION The present study demonstrated the correlation of APOAI with T cell subsets and IFN-γ in CAD. These results provided novel information for the regulatory action between APOAI and T cell subsets and inflammatory immunity in CAD.
Collapse
Affiliation(s)
- Xinlin Xiong
- Department of cardiologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
- Department of cardiologyClinical Medical College& Affiliated Hospital of Chengdu UniversityChengdu citySichuan ProvincePeople's Republic of China
| | - Zonggang Duan
- Department of cardiologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
| | - Haiyan Zhou
- Department of cardiologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
| | - Guangwei Huang
- Department of cardiologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
| | - Li Niu
- Department of cardiologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
| | - Zhenhua Luo
- Department of Central Lab, Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's HospitalThe Affiliated People's Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
- Guizhou University School of MedicineGuiyang cityGuizhou ProvincePeople's Republic of China
| | - Wei Li
- Department of cardiologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyang cityGuizhou ProvincePeople's Republic of China
| |
Collapse
|
21
|
A Study on Multiple Facets of Apolipoprotein A1 Milano. Appl Biochem Biotechnol 2023:10.1007/s12010-023-04330-2. [PMID: 36689166 DOI: 10.1007/s12010-023-04330-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/24/2023]
Abstract
For several strategies formulated to prevent atherosclerosis, Apolipoprotein A1 Milano (ApoA1M) remains a prime target. ApoA1M has been reported to have greater efficiency in reducing the incidence of coronary artery diseases. Furthermore, recombinant ApoA1M based mimetic peptide exhibits comparatively greater atheroprotective potential, offers a hope in reducing the burden of atherosclerosis in in vivo model system. The aim of this review is to emphasize on some of the observed ApoA1M structural and functional effects that are clinically and therapeutically meaningful that might converge on the basic role of ApoA1M in reducing the chances of glycation assisted ailments in diabetes. We also hypothesize that the nonenzymatic glycation prone arginine amino acid of ApoA1 gets replaced with cysteine residue and the rate of ApoA1 glycation may decrease due to change substitution of amino acid. Therefore, to circumvent the effect of ApoA1M glycation, the related mechanism should be explored at the cellular and functional levels, especially in respective experimental disease model in vivo.
Collapse
|
22
|
Stadler JT, van Poppel MNM, Christoffersen C, Hill D, Wadsack C, Simmons D, Desoye G, Marsche G. Gestational Hypertension and High-Density Lipoprotein Function: An Explorative Study in Overweight/Obese Women of the DALI Cohort. Antioxidants (Basel) 2022; 12:68. [PMID: 36670930 PMCID: PMC9854490 DOI: 10.3390/antiox12010068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022] Open
Abstract
Gestational hypertension (GHTN) is associated with an increased cardiovascular risk for mothers and their offspring later in life. High-density lipoproteins (HDL) are anti-atherogenic by promoting efflux of cholesterol from macrophages and suppression of endothelial cell activation. Functional impairment of HDL in GHTN-complicated pregnancies may affect long-term health of both mothers and offspring. We studied functional parameters of maternal and neonatal HDL in 192 obese women (pre-pregnancy BMI ≥ 29), who were at high risk for GHTN. Maternal blood samples were collected longitudinally at <20 weeks, at 24−28 and 35−37 weeks of gestation. Venous cord blood was collected immediately after birth. Maternal and cord blood were used to determine functional parameters of HDL, such as HDL cholesterol efflux capacity, activity of the vaso-protective HDL-associated enzyme paraoxonase-1, and levels of the HDL-associated anti-inflammatory apolipoprotein (apo)M. In addition, we determined serum anti-oxidative capacity. Thirteen percent of the women were diagnosed with GHTN. While we found no changes in measures of HDL function in mothers with GHTN, we observed impaired HDL cholesterol efflux capacity and paraoxonase-1 activity in cord blood, while serum antioxidant capacity was increased. Of particular interest, increased maternal paraoxonase-1 activity and apoM levels in early pregnancy were associated with the risk of developing GHTN. GHTN significantly impairs HDL cholesterol efflux capacity as well as HDL PON1 activity in cord blood and could affect vascular health in offspring. Maternal paraoxonase-1 activity and apoM levels in early pregnancy associate with the risk of developing GHTN.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - M. N. M. van Poppel
- Institute of Human Movement Science, Sport and Health, University of Graz, 8010 Graz, Austria
| | - Christina Christoffersen
- Department of Biomedical Science, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Biochemistry, Rigshospitalet, University Hospital of Copenhagen, 2200 Copenhagen, Denmark
| | - David Hill
- Lawson Health Research Institute, London, ON N6C 2R5, Canada
| | - Christian Wadsack
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - David Simmons
- Macarthur Clinical School, Western Sydney University, Sydney, NSW 2560, Australia
| | - Gernot Desoye
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | | |
Collapse
|
23
|
Ghosh S, Rihan M, Ahmed S, Pande AH, Sharma SS. Immunomodulatory potential of apolipoproteins and their mimetic peptides in asthma: Current perspective. Respir Med 2022; 204:107007. [DOI: 10.1016/j.rmed.2022.107007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/03/2022] [Indexed: 10/31/2022]
|
24
|
Neutrophils to high-density lipoprotein cholesterol ratio as a new prognostic marker in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention: a retrospective study. BMC Cardiovasc Disord 2022; 22:434. [PMID: 36199038 PMCID: PMC9533505 DOI: 10.1186/s12872-022-02870-9] [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: 03/15/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neutrophils and high-density lipoprotein cholesterol (HDL-c) play critical roles in the pathogenesis of acute myocardial infarction. We aimed to investigate the value of neutrophils count to high-density lipoprotein cholesterol ratio (NHR) in predicting occurrence of in-hospital adverse events in ST-segment elevation myocardial infarction (STEMI) patients treated with primary percutaneous coronary intervention (PPCI). METHODS We retrospectively analyzed 532 patients who had been diagnosed with acute STEMI and treated with PPCI. Demographic and clinical data, admission laboratory parameters and NHR values were recorded. Major adverse cardiac events (MACE) were defined as stent thrombosis, cardiac rupture, cardiac arrest, ventricular aneurysm, malignant arrhythmia and cardiac death. Based on the receiver operating characteristic (ROC) analysis, all patients were divided into 2 groups based on the cut-off NHR value (NHR ≤ 11.28, NHR > 11.28). Cox regression analyses and the Kaplan-Meier survival curve were used to assess the prognostic ability of NHR in in-hospital MACE. RESULTS MACE was observed in 72 patients (13.5%) during in-hospital follow-up. NHR was significantly higher in MACE group compared to MACE-free group (10.93 [6.26-13.97] vs. 8.13 [5.89-11.16]; P = 0.001). The incidence of in-hospital MACE was significantly higher in the NHR > 11.28 group than in NHR ≤ 11.28 group (24.8% vs. 9.6%; P < 0.001). In multivariable Cox regression analyses, ALT, Killip III-IV and increased NHR (hazard ratio, 2.211; 95% confidence interval,1.092-4.479; P = 0.027) were identified as independent predictive factors of occurrence of in-hospital MACE. Higher NHR group had worse cumulative survival compared with the lower group. CONCLUSIONS NHR value on admission, which is an easily calculated and universally available maker, may be useful in in-hospital risk classification of STEMI patients undergoing PPCI.
Collapse
|
25
|
Stadler JT, Mangge H, Rani A, Curcic P, Herrmann M, Prüller F, Marsche G. Low HDL Cholesterol Efflux Capacity Indicates a Fatal Course of COVID-19. Antioxidants (Basel) 2022; 11:1858. [PMID: 36290581 PMCID: PMC9598682 DOI: 10.3390/antiox11101858] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/02/2022] Open
Abstract
Plasma membrane cholesterol is required for proper trafficking and localization of receptors that facilitate severe acute respiratory syndrome coronavirus 2 infection. High-density lipoproteins (HDL) mobilize plasma membrane cholesterol, and HDL-cholesterol levels are associated with the severity of COVID-19 disease and mortality. However, HDL-cholesterol levels poorly reflect the function of this complex family of particles, and a detailed assessment of COVID-19-associated changes in HDL functionality and its prognostic value is lacking. In the present study, we assessed HDL cholesterol efflux capacity, HDL anti-inflammatory and antioxidant properties, and changes in HDL composition and metabolism in COVID-19 (n = 48) and non-COVID pneumonia patients (n = 32). COVID-19 infection markedly reduced the activity of lecithin-cholesteryl-acyltransferase and functional parameters of HDL, such as the cholesterol efflux capacity, arylesterase activity of paraoxonase 1, and anti-oxidative capacity of apoB-depleted serum when compared to non-COVID pneumonia at baseline, paralleled by markedly reduced levels of HDL-cholesterol. Of particular interest, low HDL cholesterol efflux capacity was associated with increased mortality risk in COVID-19 patients, independent of HDL-C levels. Our results highlight profound effects of COVID-19 infection on HDL function, metabolism, and composition. Low HDL cholesterol efflux capacity indicates a fatal course of COVID-19, independent of HDL-cholesterol levels.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Pero Curcic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Florian Prüller
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| |
Collapse
|
26
|
Li Y, Shan J. Study on the correlation between high density lipoprotein and lower extremities deep venous thrombosis in patients undergoing hip arthroplasty. Phlebology 2022; 37:516-521. [PMID: 35575216 DOI: 10.1177/02683555221090309] [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: 11/15/2022]
Abstract
OBJECTIVE To explore the relationship between high density lipoprotein (HDL) and lower extremities deep venous thrombosis (DVT) in patients undergoing hip arthroplasty. METHODS A total of 348 patients undergoing hip arthroplasty in our hospital were enrolled, and divided into observation (n = 154, 44.25%) and control (n = 194, 55.75%) groups according to the occurrence of lower extremities DVT. The presence of DVT was assessed 1 day before surgery and routinely every 2 days after surgery. The factors of DVT were analyzed by single factor analysis, multivariate logistic regression analysis, and Pearson correlation. RESULTS The age and body mass index in the observation group were significantly higher (p = .045, p = .041, respectively), while HDL-C was significantly lower (p = .032) than the control group. Increase age, high BMI, low apolipoprotein-A1 level and low HDL-C level were risk factors for lower extremities DVT. The mean HDL-C in the observation and control groups was 0.91 ± 0.27 and 1.19 ± 0.37, respectively, the adjusted odds ratio was 1.050; 95% CI 1.010-1.092, p = .014. CONCLUSION Elderly patients with high BMI and low HDL-C level undergoing hip arthroplasty are at risk of lower extremities DVT, and should be paid attention to clinically.
Collapse
Affiliation(s)
- Yong Li
- Department of Orthopaedic, 117858Dongyang People's Hospital, Dongyang, China
| | - Junbiao Shan
- Department of Orthopaedic, 117858Dongyang People's Hospital, Dongyang, China
| |
Collapse
|
27
|
Serum Proteomic Profile of Asthmatic Patients after Six Months of Benralizumab and Mepolizumab Treatment. Biomedicines 2022; 10:biomedicines10040761. [PMID: 35453511 PMCID: PMC9027545 DOI: 10.3390/biomedicines10040761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/05/2022] [Accepted: 03/22/2022] [Indexed: 12/02/2022] Open
Abstract
Severe eosinophilic asthma is characterized by chronic airway inflammation, oxidative stress, and elevated proinflammatory cytokines, especially IL-5. Mepolizumab and benralizumab are both humanized IgG antibodies directed against IL-5 signaling, directly acting on eosinophils count. Together with the complexity of severe asthma classification and patient selection for the targeted treatment, there is also the urgency to clarify the follow-up of therapy to identify biomarkers, in addition to eosinophils, for the optimal duration of treatment, persistence of effectiveness, and safety. To this purpose, here we performed a follow-up study using differential proteomic analysis on serum samples after 1 and 6 months of both therapies and sera from healthy patients. Statistical analysis by PCA and heatmap analyses were performed, and identified proteins were used for enrichment analysis by MetaCore software. The analysis highlighted 82 differences among all considered conditions. In particular, 30 referred to benralizumab time point (T0, T1B, T6B) and 24 to mepolizumab time point (T0, T1M, T6M) analyses. t-SNE and heatmap analyses evidence that the differential serum protein profile at 6 months of both treatments is more similar to that of the healthy subjects. Among the identified proteins, APOAI, APOC-II, and APOC-III are upregulated principally after 6 months of benralizumab treatment, plasminogen is upregulated after 6 months of both treatments and ceruloplasmin, upregulated already after 1 month of benralizumab, becoming higher after 6 months of mepolizumab. Using enrichment analysis, identified proteins were related to lipid metabolism and transport, blood coagulation, and ECM remodeling.
Collapse
|
28
|
Understanding Myeloperoxidase-Induced Damage to HDL Structure and Function in the Vessel Wall: Implications for HDL-Based Therapies. Antioxidants (Basel) 2022; 11:antiox11030556. [PMID: 35326206 PMCID: PMC8944857 DOI: 10.3390/antiox11030556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerosis is a disease of increased oxidative stress characterized by protein and lipid modifications in the vessel wall. One important oxidative pathway involves reactive intermediates generated by myeloperoxidase (MPO), an enzyme present mainly in neutrophils and monocytes. Tandem MS analysis identified MPO as a component of lesion derived high-density lipoprotein (HDL), showing that the two interact in the arterial wall. MPO modifies apolipoprotein A1 (apoA-I), paraoxonase 1 and certain HDL-associated phospholipids in human atheroma. HDL isolated from atherosclerotic plaques depicts extensive MPO mediated posttranslational modifications, including oxidation of tryptophan, tyrosine and methionine residues, and carbamylation of lysine residues. In addition, HDL associated plasmalogens are targeted by MPO, generating 2-chlorohexadecanal, a pro-inflammatory and endothelial barrier disrupting lipid that suppresses endothelial nitric oxide formation. Lesion derived HDL is predominantly lipid-depleted and cross-linked and exhibits a nearly 90% reduction in lecithin-cholesterol acyltransferase activity and cholesterol efflux capacity. Here we provide a current update of the pathophysiological consequences of MPO-induced changes in the structure and function of HDL and discuss possible therapeutic implications and options. Preclinical studies with a fully functional apoA-I variant with pronounced resistance to oxidative inactivation by MPO-generated oxidants are currently ongoing. Understanding the relationships between pathophysiological processes that affect the molecular composition and function of HDL and associated diseases is central to the future use of HDL in diagnostics, therapy, and ultimately disease management.
Collapse
|
29
|
Wang N, Liu H, Tian M, Liang J, Sun W, Zhang L, Pei L, Liu K, Sun S, Wu J, Gao Y, Xu Y, Wang Y, Song B. A Nomogram That Includes Neutrophils and High-Density Lipoprotein Cholesterol Can Predict the Prognosis of Acute Ischaemic Stroke. Front Neurol 2022; 13:827279. [PMID: 35280284 PMCID: PMC8914087 DOI: 10.3389/fneur.2022.827279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Lipids are implicated in inflammatory responses affecting acute ischaemic stroke prognosis. Therefore, we aimed to develop a predictive model that considers neutrophils and high-density lipoprotein cholesterol to predict its prognosis. This prospective study enrolled patients with acute ischaemic stroke within 24 h of onset between January 2015 and December 2017. The main outcome was a modified Rankin Scale score ≥3 at the 90th day of follow-up. Patients were divided into training and testing sets. The training set was divided into four states according to the median of neutrophils and high-density lipoprotein cholesterol levels in all patients. Through binary logistic regression analysis, the relationship between factors and prognosis was determined. A nomogram based on the results was developed; its predictive value was evaluated through internal and external validations. Altogether, 1,090 patients were enrolled with 872 (80%) and 218 (20%) in the training and testing sets, respectively. In the training set, the major outcomes occurred in 24 (10.4%), 24 (11.6%), 37 (17.2%), and 49 (22.3%) in states 1–4, respectively (P = 0.002). Validation of calibration and decision curve analyses showed that the nomogram showed better performance. The internal and external testing set receiver operating characteristics verified the predictive value [area under the curve = 0.794 (0.753–0.834), P < 0.001, and area under the curve = 0.973 (0.954–0.992), P < 0.001, respectively]. A nomogram that includes neutrophils and high-density lipoprotein cholesterol can predict the prognosis of acute ischaemic stroke, thus providing us with an effective visualization tool.
Collapse
Affiliation(s)
- Nan Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongbing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengke Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenxian Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luyang Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lulu Pei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shilei Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yilong Wang
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Bo Song
| |
Collapse
|
30
|
Bezsonov EE, Sobenin IA, Orekhov AN. Lipids and Lipoproteins in Health and Disease. Biomedicines 2021; 10:biomedicines10010087. [PMID: 35052767 PMCID: PMC8773467 DOI: 10.3390/biomedicines10010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- Evgeny E. Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia; (I.A.S.); (A.N.O.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology (A. P. Avtsyn Research Institute of Human Morphology), 3 Tsyurupa Street, 117418 Moscow, Russia
- Department of Biology and General Genetics, I. M. Sechenov First Moscow State Medical University (Sechenov University), 8 Izmailovsky Boulevard, 105043 Moscow, Russia
- Correspondence:
| | - Igor A. Sobenin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia; (I.A.S.); (A.N.O.)
- National Medical Research Center of Cardiology, Laboratory of Medical Genetics, Institute of Experimental Cardiology, 15a 3rd Cherepkovskaya Street, 121552 Moscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia; (I.A.S.); (A.N.O.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology (A. P. Avtsyn Research Institute of Human Morphology), 3 Tsyurupa Street, 117418 Moscow, Russia
| |
Collapse
|
31
|
Stadler JT, Marsche G. Dietary Strategies to Improve Cardiovascular Health: Focus on Increasing High-Density Lipoprotein Functionality. Front Nutr 2021; 8:761170. [PMID: 34881279 PMCID: PMC8646038 DOI: 10.3389/fnut.2021.761170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is one of the leading causes of morbidity and mortality worldwide, with increasing incidence. A cornerstone of cardiovascular disease prevention is lifestyle modification through dietary changes to influence various risk factors such as obesity, hypertension and diabetes. The effects of diet on cardiovascular health are complex. Some dietary components and metabolites directly affect the composition and structure of high-density lipoproteins (HDL) and increase anti-inflammatory and vasoprotective properties. HDLs are composed of distinct subpopulations of particles of varying size and composition that have several dynamic and context-dependent functions. The identification of potential dietary components that improve HDL functionality is currently an important research goal. One of the best-studied diets for cardiovascular health is the Mediterranean diet, consisting of fish, olive oil, fruits, vegetables, whole grains, legumes/nuts, and moderate consumption of alcohol, most commonly red wine. The Mediterranean diet, especially when supplemented with extra virgin olive oil rich in phenolic compounds, has been shown to markedly improve metrics of HDL functionality and reduce the burden, or even prevent the development of cardiovascular disease. Particularly, the phenolic compounds of extra virgin olive oil seem to exert the significant positive effects on HDL function. Moreover, supplementation of anthocyanins as well as antioxidants such as lycopene or the omega-3 fatty acid eicosapentaenoic acid improve parameters of HDL function. In this review, we aim to highlight recent discoveries on beneficial dietary patterns as well as nutritional components and their effects on cardiovascular health, focusing on HDL function.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
32
|
Luo F, Zeng KM, Cao JX, Zhou T, Lin SX, Ma WJ, Yang YP, Zhang ZH, Lu FT, Huang Y, Zhao HY, Zhang L. Predictive value of a reduction in the level of high-density lipoprotein-cholesterol in patients with non-small-cell lung cancer undergoing radical resection and adjuvant chemotherapy: a retrospective observational study. Lipids Health Dis 2021; 20:109. [PMID: 34544437 PMCID: PMC8454045 DOI: 10.1186/s12944-021-01538-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/31/2021] [Indexed: 12/25/2022] Open
Abstract
Background Cancer patients often exhibit chemotherapy-associated changes in serum lipid profiles, however, their prognostic value before and after adjuvant chemotherapy on survival among non-small-cell lung cancer (NSCLC) patients is unknown. Methods NSCLC patients undergoing radical resection and subsequent adjuvant chemotherapy from 2013 to 2017 at Sun Yat-sen University Cancer Center were retrospectively reviewed. Fasted serum lipid levels were measured before and after chemotherapy. The optimal lipid cut-off values at baseline and fluctuation were determined using X-tile™. The fluctuations in serum lipid levels and disease-free survival (DFS) were assessed. Results Serum cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein-cholesterol (HDL-C), triglyceride, apolipoprotein (Apo) A-I, and ApoB all significantly increased after adjuvant chemotherapy. X-tile determined 1.52 mmol/L of HDL-C and 0.74 g/L of ApoB as the optimal cut-off values before chemotherapy. Patients with HDL-C ≥ 1.52 mmol/L (median DFS: not reached vs. 26.30 months, P = 0.0005) and a decreased HDL-C level after adjuvant chemotherapy (median DFS: 80.43 vs. 26.12 months, P = 0.0204) had a longer DFS. An HDL-C level that increased by ≥ 0.32 mmol/L after chemotherapy indicated a worse DFS. A high baseline ApoB level were associated with a superior DFS. In the univariate analysis and the multivariate Cox analyses, a high baseline HDL-C level and a HDL-C reduction after adjuvant chemotherapy were independent indicators for superior DFS. High baseline HDL-C was related to N0-1 stage (χ2 = 6.413, P = 0.011), and HDL-C fluctuation was significantly correlated with specific chemotherapy regimens (χ2 = 5.002, P = 0.025). Conclusions Adjuvant chemotherapy increased various lipid levels in resected NSCLC patients. A higher HDL-C level before chemotherapy and a reduced HDL-C level after adjuvant chemotherapy were independent predictors of longer DFS in patients with curable NSCLC.
Collapse
Affiliation(s)
- Fan Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Kang-Mei Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China.
| | - Jia-Xin Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Ting Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Su-Xia Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Wen-Juan Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Yun-Peng Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Zhong-Han Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Fei-Teng Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Yan Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China
| | - Hong-Yun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Clinical Research, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China.
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Guangdong Esophageal Cancer Institute, Sun Yat- sen University Cancer Center, 510060, Guangzhou, People's Republic of China.
| |
Collapse
|
33
|
von Eckardstein A. High Density Lipoproteins: Is There a Comeback as a Therapeutic Target? Handb Exp Pharmacol 2021; 270:157-200. [PMID: 34463854 DOI: 10.1007/164_2021_536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Low plasma levels of High Density Lipoprotein (HDL) cholesterol (HDL-C) are associated with increased risks of atherosclerotic cardiovascular disease (ASCVD). In cell culture and animal models, HDL particles exert multiple potentially anti-atherogenic effects. However, drugs increasing HDL-C have failed to prevent cardiovascular endpoints. Mendelian Randomization studies neither found any genetic causality for the associations of HDL-C levels with differences in cardiovascular risk. Therefore, the causal role and, hence, utility as a therapeutic target of HDL has been questioned. However, the biomarker "HDL-C" as well as the interpretation of previous data has several important limitations: First, the inverse relationship of HDL-C with risk of ASCVD is neither linear nor continuous. Hence, neither the-higher-the-better strategies of previous drug developments nor previous linear cause-effect relationships assuming Mendelian randomization approaches appear appropriate. Second, most of the drugs previously tested do not target HDL metabolism specifically so that the futile trials question the clinical utility of the investigated drugs rather than the causal role of HDL in ASCVD. Third, the cholesterol of HDL measured as HDL-C neither exerts nor reports any HDL function. Comprehensive knowledge of structure-function-disease relationships of HDL particles and associated molecules will be a pre-requisite, to test them for their physiological and pathogenic relevance and exploit them for the diagnostic and therapeutic management of individuals at HDL-associated risk of ASCVD but also other diseases, for example diabetes, chronic kidney disease, infections, autoimmune and neurodegenerative diseases.
Collapse
Affiliation(s)
- Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|