1
|
Wang Z, Yin Z, Sun G, Zhang D, Zhang J. Genetic evidence for the liver-brain axis: lipid metabolism and neurodegenerative disease risk. Lipids Health Dis 2025; 24:41. [PMID: 39923073 PMCID: PMC11806572 DOI: 10.1186/s12944-025-02455-3] [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: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND The liver‒brain axis is critical in neurodegenerative diseases (NDs), with lipid metabolism influencing neuroinflammation and microglial function. A systematic investigation of the genetic relationship between lipid metabolism abnormalities and ND, namely, Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), is lacking. To assess potential causal links between ND and six lipid parameters, two-sample Mendelian randomization (MR) was used. METHODS Large-scale European ancestry GWAS data for lipid parameters and ND (AD, ALS, PD, and MS) were used. Genetic variants demonstrating significant correlations (P < 5 × 10-8) with lipid metabolism parameters were identified and employed as instrumental variables (IVs) after proper validation. The research incorporated UK Biobank genomic data to examine associations between genetic variants and lipid metabolism parameters. The analysis included primary MR, sensitivity analyses, and multivariable MR, which considered potential mediators. RESULTS MR via the inverse-variance weighted method revealed causal effects of cholesterol (CHOL, OR = 1.10, 95% CI: 1.03-1.18, P = 4.23 × 10⁻3) and low-density lipoprotein cholesterol (LDLC, OR = 1.10, 95% CI: 1.03-1.17, P = 3.28 × 10⁻3) on the risk of ALS, which were validated across multiple methods. Potential correlations were observed between ApoB and ALS and inversely correlated with AD, whereas no significant associations were found for PD or MS. CHOL and LDLC associations with ALS demonstrated no significant heterogeneity or pleiotropy, supporting their reliability. CONCLUSIONS Higher CHOL and LDLC levels were associated with increased ALS risk, suggesting a potential causal link, and supporting the liver‒brain axis hypothesis in ND. Current genetic evidence does not support a significant role for lipid metabolism in PD and MS etiology, suggesting the relationship between lipid metabolism and other NDs may be more complex and warrants further investigation.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zixiao Yin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China
| | - Guangyong Sun
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dong Zhang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China.
| |
Collapse
|
2
|
Chen L, Mei-Ling Tan K, Leow MKS, Tan KH, Yen Chan JK, Chan SY, Chong YS, Gluckman PD, Eriksson JG, Wenk MR, Mir SA. Characterisation of pregnancy-induced alterations in apolipoproteins and their associations with maternal metabolic risk factors and offspring birth outcomes: a preconception and longitudinal cohort study. EBioMedicine 2025; 112:105562. [PMID: 39842285 PMCID: PMC11794176 DOI: 10.1016/j.ebiom.2025.105562] [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: 08/08/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Apolipoproteins as an integral part of lipoproteins are crucial for the transport and metabolism of lipids. However, there is a lack of longitudinal studies to quantify the concentrations of maternal apolipoproteins from preconception to postpartum and their associations with maternal metabolic health and offspring birth outcomes. METHODS Quantification of apolipoproteins was performed on maternal plasma samples (N = 243 trios) collected at preconception, 26-28 weeks' pregnancy, and three months postpartum in the Singapore PREconception Study of long-Term maternal and child Outcomes (S-PRESTO) cohort study. Linear regression models and network analysis were implemented to investigate the association of apolipoproteins with maternal genetic variants, biochemical measures, metabolic risk factors, and offspring birth outcomes. FINDINGS The concentrations of ApoC-III, ApoB and ApoL1 substantially increased in pregnancy compared to preconception and postpartum. Genome-wide association studies (GWAS) identified multiple single-nucleotide polymorphisms (SNPs) associated with plasma apolipoproteins (P < 5.00E-08), including APOE-rs7412 for ApoE, LPA-rs56393506 for Apo(a), APOM-rs707921 for ApoM, ABCC4-rs117797426 for ApoJ, THSD7B-rs575613 for ApoA-II, and LOC102724443-rs140433245 for ApoA-IV. Plasma apolipoproteins were strongly associated with biochemical measures including lipidomic profiles, lipoprotein features and fat-soluble vitamins, as well as metabolic risk factors including glycaemic traits, liver enzymes, inflammatory markers, albumin, and blood pressure. Integrative network analysis of apolipoproteins and their correlates/determinants revealed both shared and specific associations, with the strongest relationships observed among apolipoproteins, cholesterol, triglycerides, alpha tocopherol, and GlycA (Padj < 0.05). Higher maternal ApoC-I and ApoC-III concentrations at preconception were significantly associated with shorter gestational age of the offspring. INTERPRETATION We describe the longitudinal landscape of maternal circulating apolipoproteins from preconception to postpartum and their associations with maternal metabolic risk factors and offspring birth outcomes. This multi-omics characterisation of biochemical correlates and genetic determinants of maternal apolipoproteins will deepen our understanding of the molecular basis of metabolic flexibility in expectant mothers, leading to better assessment of pregnancy-related outcomes. FUNDING This research was supported by the Singapore National Research Foundation under its Translational and Clinical Research (TCR) Flagship Programme and administered by the Singapore Ministry of Health's National Medical Research Council (NMRC), Singapore- NMRC/TCR/004-NUS/2008; NMRC/TCR/012-NUHS/2014. The Singapore Lipidomics Incubator (SLING) is supported by grants from the National University of Singapore via the Life Sciences Institute, the National Research Foundation (NRF, NRFI2015-05 and NRFSBP-P4) and A∗STAR IAF-ICP I1901E0040. Additional funding is provided by Institute for Human Development and Potential (IHDP)-Agency for Science, Technology and Research (A∗STAR), Singapore.
Collapse
Affiliation(s)
- Li Chen
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Karen Mei-Ling Tan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Melvin Khee-Shing Leow
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, Singapore; KK Women's and Children's Hospital, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Duke-NUS Medical School, Singapore, Singapore; KK Women's and Children's Hospital, Singapore, Singapore
| | - Shiao-Yng Chan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Folkhalsan Research Center, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar; Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Masuda D, Okada T, Sairyou M, Takafuji K, Ohama T, Koseki M, Nishida M, Sakata Y, Yamashita S. Proteomic Analysis of Human Chylomicron Remnants Isolated by Apolipoprotein B-48 Immunoprecipitation. J Atheroscler Thromb 2025; 32:226-238. [PMID: 39085140 PMCID: PMC11802255 DOI: 10.5551/jat.64920] [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: 02/21/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
AIM Postprandial hypertriglyceridemia (PHTG) is an independent risk factor for coronary heart diseases. PHTG exhibits accumulation of apoB-48 containing chylomicron remnants (CM-Rs) and apoB-100 containing VLDL remnants (VLDL-Rs), which are both known to be atherogenic. However, unlike VLDL-Rs, structural and functional characterization of CM-Rs remains to be elucidated due to challenges in separating CM-Rs from VLDL-Rs. Recently, we successfully isolated CM-Rs and VLDL-Rs utilizing anti-apoB-48 or apoB-100 specific antibodies. This study aimed to characterize the proteome of CM-Rs along with that of VLDL-Rs. METHODS Eight healthy subjects were enrolled. Venous blood was drawn 3 hours after high-fat-containing meals. We isolated CM-Rs and VLDL-Rs from sera through combination of ultracentrifugation and immunoprecipitation using apoB-48 or apoB-100 specific antibodies, followed by shotgun proteomic analysis. RESULTS We identified 42 CM-Rs or VLDL-Rs-associated proteins, including 11 potential newly identified proteins such as platelet basic protein (PPBP) and platelet factor 4, which are chemokines secreted from platelets. ApoA-I, apoA-IV, and clusterin, which are also known as HDL-associated proteins, were significantly more abundant in CM-Rs. Interestingly, apoC-I, which reduces the activity of lipoprotein lipase and eventually inhibits catabolism of remnant proteins, was also more abundant in CM-Rs. Moreover, we identified proteins involved in complement regulation such as complement C3 and vitronectin, and those involved in acute-phase response such as PPBP, serum amyloid A protein 2, and protein S100-A8, in both CM-Rs and VLDL-Rs. CONCLUSIONS We have firstly characterized the proteome of CM-Rs. These findings may provide an explanation for the atherogenic properties of CM-Rs.
Collapse
Affiliation(s)
- Daisaku Masuda
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Cardiology, Rinku General Medical Center, Izumisano, Osaka, Japan
| | - Takeshi Okada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Community Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masami Sairyou
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Takafuji
- Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tohru Ohama
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masahiro Koseki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Nishida
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Health Care Division, Health and Counseling Center, Osaka University, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shizuya Yamashita
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Community Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Cardiology, Rinku General Medical Center, Izumisano, Osaka, Japan
| |
Collapse
|
4
|
Ramírez-Melo LM, Estrada-Luna D, Rubio-Ruiz ME, Castañeda-Ovando A, Fernández-Martínez E, Jiménez-Osorio AS, Pérez-Méndez Ó, Carreón-Torres E. Relevance of Lipoprotein Composition in Endothelial Dysfunction and the Development of Hypertension. Int J Mol Sci 2025; 26:1125. [PMID: 39940892 PMCID: PMC11817739 DOI: 10.3390/ijms26031125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Endothelial dysfunction and chronic inflammation are determining factors in the development and progression of chronic degenerative diseases, such as hypertension and atherosclerosis. Among the shared pathophysiological characteristics of these two diseases is a metabolic disorder of lipids and lipoproteins. Therefore, the contents and quality of the lipids and proteins of lipoproteins become the targets of therapeutic objective. One of the stages of lipoprotein formation occurs through the incorporation of dietary lipids by enterocytes into the chylomicrons. Consequently, the composition, structure, and especially the properties of lipoproteins could be modified through the intake of bioactive compounds. The objective of this review is to describe the roles of the different lipid and protein components of lipoproteins and their receptors in endothelial dysfunction and the development of hypertension. In addition, we review the use of some non-pharmacological treatments that could improve endothelial function and/or prevent endothelial damage. The reviewed information contributes to the understanding of lipoproteins as vehicles of regulatory factors involved in the modulation of inflammatory and hemostatic processes, the attenuation of oxidative stress, and the neutralization of toxins, rather than only cholesterol and phospholipid transporters. For this review, a bibliographic search was carried out in different online metabases.
Collapse
Affiliation(s)
- Lisette Monsibaez Ramírez-Melo
- Nutrition Academic Area Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico;
| | - Diego Estrada-Luna
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Tlalpan, Mexico City 14080, Mexico;
| | - Araceli Castañeda-Ovando
- Chemistry Academic Area, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Eduardo Fernández-Martínez
- Medicine Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Angélica Saraí Jiménez-Osorio
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
- Tecnológico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
| |
Collapse
|
5
|
Németh K, Kestecher BM, Ghosal S, Bodnár BR, Kittel Á, Hambalkó S, Kovácsházi C, Giricz Z, Ferdinandy P, Osteikoetxea X, Burkhardt R, Buzas EI, Orsó E. Therapeutic and pharmacological applications of extracellular vesicles and lipoproteins. Br J Pharmacol 2024; 181:4733-4749. [PMID: 39491825 DOI: 10.1111/bph.17336] [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: 04/12/2024] [Revised: 06/15/2024] [Accepted: 07/13/2024] [Indexed: 11/05/2024] Open
Abstract
In recent years, various approaches have been undertaken to eliminate lipoproteins co-isolated with extracellular vesicles, as they were initially regarded as contaminating entities. However, novel discoveries are reshaping our perspective. In body fluids, these distinct particles not only co-exist, but also interactions between them are likely to occur. Extracellular vesicles and lipoproteins can associate with each other, share cargo, influence each other's functions, and jointly have a role in the pathomechanisms of diseases. Additionally, their association carries important implications for therapeutic and pharmacological aspects of lipid-lowering strategies. Extracellular vesicles and lipoprotein particles may have roles in the elimination of each other from the circulation. The objective of this minireview is to delve into these aspects. Here, we show that under certain physiological and pathological conditions, extracellular vesicles and lipoproteins are 'partners' rather than 'strangers' or 'rivals'.
Collapse
Affiliation(s)
- Krisztina Németh
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
| | - Brachyahu M Kestecher
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Sayam Ghosal
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Bernadett R Bodnár
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ágnes Kittel
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HUN-REN, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Xabier Osteikoetxea
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Edit I Buzas
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Evelyn Orsó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Bhale AS, Meilhac O, d'Hellencourt CL, Vijayalakshmi MA, Venkataraman K. Cholesterol transport and beyond: Illuminating the versatile functions of HDL apolipoproteins through structural insights and functional implications. Biofactors 2024; 50:922-956. [PMID: 38661230 DOI: 10.1002/biof.2057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
High-density lipoproteins (HDLs) play a vital role in lipid metabolism and cardiovascular health, as they are intricately involved in cholesterol transport and inflammation modulation. The proteome of HDL particles is indeed complex and distinct from other components in the bloodstream. Proteomics studies have identified nearly 285 different proteins associated with HDL; however, this review focuses more on the 15 or so traditionally named "apo" lipoproteins. Important lipid metabolizing enzymes closely working with the apolipoproteins are also discussed. Apolipoproteins stand out for their integral role in HDL stability, structure, function, and metabolism. The unique structure and functions of each apolipoprotein influence important processes such as inflammation regulation and lipid metabolism. These interactions also shape the stability and performance of HDL particles. HDLs apolipoproteins have multifaceted roles beyond cardiovascular diseases (CVDs) and are involved in various physiological processes and disease states. Therefore, a detailed exploration of these apolipoproteins can offer valuable insights into potential diagnostic markers and therapeutic targets. This comprehensive review article aims to provide an in-depth understanding of HDL apolipoproteins, highlighting their distinct structures, functions, and contributions to various physiological processes. Exploiting this knowledge holds great potential for improving HDL function, enhancing cholesterol efflux, and modulating inflammatory processes, ultimately benefiting individuals by limiting the risks associated with CVDs and other inflammation-based pathologies. Understanding the nature of all 15 apolipoproteins expands our knowledge of HDL metabolism, sheds light on their pathological implications, and paves the way for advancements in the diagnosis, prevention, and treatment of lipid and inflammatory-related disorders.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | - Christian Lefebvre d'Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | | | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
7
|
López-Valencia L, Moya M, Escudero B, García-Bueno B, Orio L. Bacterial lipopolysaccharide forms aggregates with apolipoproteins in male and female rat brains after ethanol binges. J Lipid Res 2024; 65:100509. [PMID: 38295984 PMCID: PMC10907226 DOI: 10.1016/j.jlr.2024.100509] [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: 08/09/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 03/01/2024] Open
Abstract
Alcohol binge drinking allows the translocation of bacterial lipopolysaccharide (LPS) from the gut to the blood, which activates the peripheral immune system with consequences in neuroinflammation. A possible access/direct signaling of LPS to/in the brain has not yet been described under alcohol abuse conditions. Apolipoproteins are compounds altered by alcohol with high affinity to LPS which may be involved in its transport to the brain or in its elimination. Here, we explored the expression of small components of LPS, in its free form or bound to apolipoproteins, in the brain of female and male rats exposed to alcohol binges. Animals received ethanol oral gavages (3 g/kg every 8 h) for 4 days. LPS or its components (Lipid A and core), LPS-binding protein, corticosterone, lipoproteins (HDL, LDL), apolipoproteins (ApoAI, ApoB, and ApoE), and their receptors were measured in plasma and/or in nonperfused prefrontal cortex (PFC) and cerebellum. Brain LipidA-apolipoprotein aggregates were determined by Western blotting and confirmed by co-immunoprecipitation. In animals exposed to alcohol binges: 1) plasma LPS-binding protein was elevated in both sexes; 2) females showed elevations in plasma ApoAI and corticosterone levels; 3) Lipid A formed aggregates with ApoAI in the female PFC and with ApoB in males, the latter showing Toll-like receptor 4 upregulation in PFC but not females. These results suggest that small bacterial components are present within the brain, forming aggregates with different apolipoproteins, depending on the sex, after alcohol binge intoxications. Results may have implications for the crosstalk between alcohol, LPS, and neuroinflammation.
Collapse
Affiliation(s)
- L López-Valencia
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain; Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Neuroscience and Mental Health, Madrid, Spain
| | - M Moya
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain
| | - B Escudero
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain; Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Neuroscience and Mental Health, Madrid, Spain
| | - B García-Bueno
- Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Neuroscience and Mental Health, Madrid, Spain; Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain; Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - L Orio
- Department of Psychobiology and Behavioral Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Pozuelo de Alarcón, Spain; Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre (imas12), Neuroscience and Mental Health, Madrid, Spain; RIAPAd: Research network in primary care in addictions ('Red de investigación en atención primaria en adicciones'), Spain.
| |
Collapse
|
8
|
Nieddu G, Formato M, Lepedda AJ. Searching for Atherosclerosis Biomarkers by Proteomics: A Focus on Lesion Pathogenesis and Vulnerability. Int J Mol Sci 2023; 24:15175. [PMID: 37894856 PMCID: PMC10607641 DOI: 10.3390/ijms242015175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Plaque rupture and thrombosis are the most important clinical complications in the pathogenesis of stroke, coronary arteries, and peripheral vascular diseases. The identification of early biomarkers of plaque presence and susceptibility to ulceration could be of primary importance in preventing such life-threatening events. With the improvement of proteomic tools, large-scale technologies have been proven valuable in attempting to unravel pathways of atherosclerotic degeneration and identifying new circulating markers to be utilized either as early diagnostic traits or as targets for new drug therapies. To address these issues, different matrices of human origin, such as vascular cells, arterial tissues, plasma, and urine, have been investigated. Besides, proteomics was also applied to experimental atherosclerosis in order to unveil significant insights into the mechanisms influencing atherogenesis. This narrative review provides an overview of the last twenty years of omics applications to the study of atherogenesis and lesion vulnerability, with particular emphasis on lipoproteomics and vascular tissue proteomics. Major issues of tissue analyses, such as plaque complexity, sampling, availability, choice of proper controls, and lipoproteins purification, will be raised, and future directions will be addressed.
Collapse
Affiliation(s)
| | | | - Antonio Junior Lepedda
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (G.N.); (M.F.); Antonio Junior Lepedda (A.J.L.)
| |
Collapse
|
9
|
Pérez-Luz S, Lalchandani J, Matamala N, Barrero MJ, Gil-Martín S, Saz SRD, Varona S, Monzón S, Cuesta I, Justo I, Marcacuzco A, Hierro L, Garfia C, Gomez-Mariano G, Janciauskiene S, Martínez-Delgado B. Quantitative Lipid Profiling Reveals Major Differences between Liver Organoids with Normal Pi*M and Deficient Pi*Z Variants of Alpha-1-antitrypsin. Int J Mol Sci 2023; 24:12472. [PMID: 37569847 PMCID: PMC10419530 DOI: 10.3390/ijms241512472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Different mutations in the SERPINA1 gene result in alpha-1 antitrypsin (AAT) deficiency and in an increased risk for the development of liver diseases. More than 90% of severe deficiency patients are homozygous for Z (Glu342Lys) mutation. This mutation causes Z-AAT polymerization and intrahepatic accumulation which can result in hepatic alterations leading to steatosis, fibrosis, cirrhosis, and/or hepatocarcinoma. We aimed to investigate lipid status in hepatocytes carrying Z and normal M alleles of the SERPINA1 gene. Hepatic organoids were developed to investigate lipid alterations. Lipid accumulation in HepG2 cells overexpressing Z-AAT, as well as in patient-derived hepatic organoids from Pi*MZ and Pi*ZZ individuals, was evaluated by Oil-Red staining in comparison to HepG2 cells expressing M-AAT and liver organoids from Pi*MM controls. Furthermore, mass spectrometry-based lipidomics analysis and transcriptomic profiling were assessed in Pi*MZ and Pi*ZZ organoids. HepG2 cells expressing Z-AAT and liver organoids from Pi*MZ and Pi*ZZ patients showed intracellular accumulation of AAT and high numbers of lipid droplets. These latter paralleled with augmented intrahepatic lipids, and in particular altered proportion of triglycerides, cholesterol esters, and cardiolipins. According to transcriptomic analysis, Pi*ZZ organoids possess many alterations in genes and cellular processes of lipid metabolism with a specific impact on the endoplasmic reticulum, mitochondria, and peroxisome dysfunction. Our data reveal a relationship between intrahepatic accumulation of Z-AAT and alterations in lipid homeostasis, which implies that liver organoids provide an excellent model to study liver diseases related to the mutation of the SERPINA1 gene.
Collapse
Affiliation(s)
- Sara Pérez-Luz
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Jaanam Lalchandani
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Nerea Matamala
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Maria Jose Barrero
- Models and Mechanisms Unit, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain;
| | - Sara Gil-Martín
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER U758, 28029 Madrid, Spain
| | - Sheila Ramos-Del Saz
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Sarai Varona
- Bioinformatics Unit, Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.V.); (S.M.); (I.C.)
| | - Sara Monzón
- Bioinformatics Unit, Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.V.); (S.M.); (I.C.)
| | - Isabel Cuesta
- Bioinformatics Unit, Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.V.); (S.M.); (I.C.)
| | - Iago Justo
- General and Digestive Surgery Department, Hospital 12 de Octubre, 28041 Madrid, Spain; (I.J.); (A.M.)
| | - Alberto Marcacuzco
- General and Digestive Surgery Department, Hospital 12 de Octubre, 28041 Madrid, Spain; (I.J.); (A.M.)
| | - Loreto Hierro
- Paediatric Hepatology Service, Research Institute of University Hospital La Paz, (IdiPAZ), 28046 Madrid, Spain;
| | - Cristina Garfia
- Digestive Department, Hospital 12 de Octubre, 28041 Madrid, Spain;
| | - Gema Gomez-Mariano
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany;
| | - Beatriz Martínez-Delgado
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER U758, 28029 Madrid, Spain
| |
Collapse
|
10
|
Song S, Wu H, Liu Y, Lan D, Jiao B, Wan S, Guo Y, Zhou D, Ding Y, Ji X, Meng R. Remote ischemic conditioning-induced hyperacute and acute responses of plasma proteome in healthy young male adults: a quantitative proteomic analysis. Chin Med J (Engl) 2023; 136:150-158. [PMID: 36848171 PMCID: PMC10106146 DOI: 10.1097/cm9.0000000000002572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Long-term remote ischemic conditioning (RIC) has been proven to be beneficial in multiple diseases, such as cerebral and cardiovascular diseases. However, the hyperacute and acute effects of a single RIC stimulus are still not clear. Quantitative proteomic analyses of plasma proteins following RIC application have been conducted in preclinical and clinical studies but exhibit high heterogeneity in results due to wide variations in experimental setups and sampling procedures. Hence, this study aimed to explore the immediate effects of RIC on plasma proteome in healthy young adults to exclude confounding factors of disease entity, such as medications and gender. METHODS Young healthy male participants were enrolled after a systematic physical examination and 6-month lifestyle observation. Individual RIC sessions included five cycles of alternative ischemia and reperfusion, each lasting for 5 min in bilateral forearms. Blood samples were collected at baseline, 5 min after RIC, and 2 h after RIC, and then samples were processed for proteomic analysis using liquid chromatography-tandem mass spectrometry method. RESULTS Proteins related to lipid metabolism (e.g., Apolipoprotein F), coagulation factors (hepatocyte growth factor activator preproprotein), members of complement cascades (mannan-binding lectin serine protease 1 isoform 2 precursor), and inflammatory responses (carboxypeptidase N catalytic chain precursor) were differentially altered at their serum levels following the RIC intervention. The most enriched pathways were protein glycosylation and complement/coagulation cascades. CONCLUSIONS One-time RIC stimulus may induce instant cellular responses like anti-inflammation, coagulation, and fibrinolysis balancing, and lipid metabolism regulation which are protective in different perspectives. Protective effects of single RIC in hyperacute and acute phases may be exploited in clinical emergency settings due to apparently beneficial alterations in plasma proteome profile. Furthermore, the beneficial effects of long-term (repeated) RIC interventions in preventing chronic cardiovascular diseases among general populations can also be expected based on our study findings.
Collapse
Affiliation(s)
- Siying Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yunhuan Liu
- Department of Neurology, Huadong Hospital, Fudan University, Shanghai 200031, China
| | - Duo Lan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Baolian Jiao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuling Wan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yibing Guo
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuchuan Ding
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
11
|
Bhale AS, Venkataraman K. Leveraging knowledge of HDLs major protein ApoA1: Structure, function, mutations, and potential therapeutics. Biomed Pharmacother 2022; 154:113634. [PMID: 36063649 DOI: 10.1016/j.biopha.2022.113634] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022] Open
Abstract
Apolipoprotein A1 (ApoA1) is a member of the Apolipoprotein family of proteins. It's a vital protein that helps in the production of high-density lipoprotein (HDL) particles, which are crucial for reverse cholesterol transport (RCT). It also has anti-inflammatory, anti-atherogenic, anti-apoptotic, and anti-thrombotic properties. These functions interact to give HDL particles their cardioprotective characteristics. ApoA1 has recently been investigated for its potential role in atherosclerosis, diabetes, neurological diseases, cancer, and certain infectious diseases. Since ApoA1's discovery, numerous mutations have been reported that affect its structural integrity and alter its function. Hence these insights have led to the development of clinically relevant peptides and synthetic reconstituted HDL (rHDL) that mimics the function of ApoA1. As a result, this review has aimed to provide an organized explanation of our understanding of the ApoA1 protein structure and its role in various essential pathways. Furthermore, we have comprehensively reviewed the important ApoA1 mutations (24 mutations) that are reported to be involved in various diseases. Finally, we've focused on the therapeutic potentials of some of the beneficial mutations, small peptides, and synthetic rHDL that are currently being researched or developed, since these will aid in the development of novel therapeutics in the future.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
12
|
Abstract
Apolipoproteins, the protein component of lipoproteins, play an important role in lipid transport, lipoprotein assembly, and receptor recognition. Apolipoproteins are glycosylated and the glycan moieties play an integral role in apolipoprotein function. Changes in apolipoprotein glycosylation correlate with several diseases manifesting in dyslipidemias. Despite their relevance in apolipoprotein function and diseases, the total glycan repertoire of most apolipoproteins remains undefined. This review summarizes the current knowledge and knowledge gaps regarding human apolipoprotein glycan composition, structure, glycosylation site, and functions. Given the relevance of glycosylation to apolipoprotein function, we expect that future studies of apolipoprotein glycosylation will contribute new understanding of disease processes and uncover relevant biomarkers and therapeutic targets. Considering these future efforts, we also provide a brief overview of current mass spectrometry based technologies that can be applied to define detailed glycan structures, site-specific compositions, and the role of emerging approaches for clinical applications in biomarker discovery and personalized medicine.
Collapse
|
13
|
Wang D, Yu B, Li Q, Guo Y, Koike T, Koike Y, Wu Q, Zhang J, Mao L, Tang X, Sun L, Lin X, Wu J, Chen YE, Peng D, Zeng R. OUP accepted manuscript. J Mol Cell Biol 2022; 14:6547772. [PMID: 35278086 PMCID: PMC9254886 DOI: 10.1093/jmcb/mjac004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 11/12/2022] Open
Abstract
Lipoprotein, especially high-density lipoprotein (HDL), particles are composed of multiple heterogeneous subgroups containing various proteins and lipids. The molecular distribution among these subgroups is closely related to cardiovascular disease (CVD). Here, we established high-resolution proteomics and lipidomics (HiPL) methods to depict the molecular profiles across lipoprotein (Lipo-HiPL) and HDL (HDL-HiPL) subgroups by optimizing the resolution of anion-exchange chromatography and comprehensive quantification of proteins and lipids on the omics level. Furthermore, based on the Pearson correlation coefficient analysis of molecular profiles across high-resolution subgroups, we achieved the relationship of proteome‒lipidome connectivity (PLC) for lipoprotein and HDL particles. By application of these methods to high-fat, high-cholesterol diet-fed rabbits and acute coronary syndrome (ACS) patients, we uncovered the delicate dynamics of the molecular profile and reconstruction of lipoprotein and HDL particles. Of note, the PLC features revealed by the HDL-HiPL method discriminated ACS from healthy individuals better than direct proteome and lipidome quantification or PLC features revealed by the Lipo-HiPL method, suggesting their potential in ACS diagnosis. Together, we established HiPL methods to trace the dynamics of the molecular profile and PLC of lipoprotein and even HDL during the development of CVD.
Collapse
Affiliation(s)
| | | | | | | | - Tomonari Koike
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yui Koike
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Qingqing Wu
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ling Mao
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiaoyu Tang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Liang Sun
- Key Laboratory of Nutrition and Metabolism, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiarui Wu
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | | | | | - Rong Zeng
- Correspondence to: Rong Zeng, E-mail:
| |
Collapse
|
14
|
Finamore F, Nieddu G, Rocchiccioli S, Spirito R, Guarino A, Formato M, Lepedda AJ. Apolipoprotein Signature of HDL and LDL from Atherosclerotic Patients in Relation with Carotid Plaque Typology: A Preliminary Report. Biomedicines 2021; 9:biomedicines9091156. [PMID: 34572342 PMCID: PMC8465382 DOI: 10.3390/biomedicines9091156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022] Open
Abstract
In the past years, it has become increasingly clear that the protein cargo of the different lipoprotein classes is largely responsible for carrying out their various functions, also in relation to pathological conditions, including atherosclerosis. Accordingly, detailed information about their apolipoprotein composition and structure may contribute to the revelation of their role in atherogenesis and the understanding of the mechanisms that lead to atherosclerotic degeneration and toward vulnerable plaque formation. With this aim, shotgun proteomics was applied to identify the apolipoprotein signatures of both high-density and low-density lipoproteins (HDL and LDL) plasma fractions purified from healthy volunteers and atherosclerotic patients with different plaque typologies who underwent carotid endarterectomy. By this approach, two proteins with potential implications in inflammatory, immune, and hemostatic pathways, namely, integrin beta-2 (P05107) and secretoglobin family 3A member 2 (Q96PL1), have been confirmed to belong to the HDL proteome. Similarly, the list of LDL-associated proteins has been enriched with 21 proteins involved in complement and coagulation cascades and the acute-phase response, which potentially double the protein species of LDL cargo. Moreover, differential expression analysis has shown protein signatures specific for patients with “hard” or “soft” plaques.
Collapse
Affiliation(s)
- Francesco Finamore
- Institute of Clinical Physiology, National Research Council, via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.F.); (S.R.)
| | - Gabriele Nieddu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy; (G.N.); (M.F.)
| | - Silvia Rocchiccioli
- Institute of Clinical Physiology, National Research Council, via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.F.); (S.R.)
| | - Rita Spirito
- Centro Cardiologico Monzino, IRCCS, via Parea 4, 20138 Milano, Italy; (R.S.); (A.G.)
| | - Anna Guarino
- Centro Cardiologico Monzino, IRCCS, via Parea 4, 20138 Milano, Italy; (R.S.); (A.G.)
| | - Marilena Formato
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy; (G.N.); (M.F.)
| | - Antonio Junior Lepedda
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy; (G.N.); (M.F.)
- Correspondence:
| |
Collapse
|
15
|
Han YH, Onufer EJ, Huang LH, Sprung RW, Davidson WS, Czepielewski RS, Wohltmann M, Sorci-Thomas MG, Warner BW, Randolph GJ. Enterically derived high-density lipoprotein restrains liver injury through the portal vein. Science 2021; 373:eabe6729. [PMID: 34437091 PMCID: PMC8478306 DOI: 10.1126/science.abe6729] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 03/16/2021] [Accepted: 06/03/2021] [Indexed: 12/30/2022]
Abstract
The biogenesis of high-density lipoprotein (HDL) requires apoA1 and the cholesterol transporter ABCA1. Although the liver generates most of the HDL in the blood, HDL synthesis also occurs in the small intestine. Here, we show that intestine-derived HDL traverses the portal vein in the HDL3 subspecies form, in complex with lipopolysaccharide (LPS)-binding protein (LBP). HDL3, but not HDL2 or low-density lipoprotein, prevented LPS binding to and inflammatory activation of liver macrophages and instead supported extracellular inactivation of LPS. In mouse models involving surgical, dietary, or alcoholic intestinal insult, loss of intestine-derived HDL worsened liver injury, whereas outcomes were improved by therapeutics that elevated and depended upon raising intestinal HDL. Thus, protection of the liver from injury in response to gut-derived LPS is a major function of intestinally synthesized HDL.
Collapse
Affiliation(s)
- Yong-Hyun Han
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Emily J Onufer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Li-Hao Huang
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert W Sprung
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA
| | - Rafael S Czepielewski
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mary Wohltmann
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mary G Sorci-Thomas
- Department of Medicine, Division of Endocrinology, Pharmacology and Toxicology, and Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brad W Warner
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gwendalyn J Randolph
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
16
|
Comparison of Plasma Lipoprotein Composition and Function in Cerebral Amyloid Angiopathy and Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9010072. [PMID: 33445800 PMCID: PMC7828227 DOI: 10.3390/biomedicines9010072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) refers to beta-amyloid (Aβ) deposition in brain vessels and is clinically the main cause of lobar intracerebral hemorrhage (ICH). Aβ can also accumulate in brain parenchyma forming neuritic plaques in Alzheimer’s disease (AD). Our study aimed to determine whether the peripheral lipid profile and lipoprotein composition are associated with cerebral beta-amyloidosis pathology and may reflect biological differences in AD and CAA. For this purpose, lipid and apolipoproteins levels were analyzed in plasma from 51 ICH-CAA patients (collected during the chronic phase of the disease), 60 AD patients, and 60 control subjects. Lipoproteins (VLDL, LDL, and HDL) were isolated and their composition and pro/antioxidant ability were determined. We observed that alterations in the lipid profile and lipoprotein composition were remarkable in the ICH-CAA group compared to control subjects, whereas the AD group presented no specific alterations compared with controls. ICH-CAA patients presented an atheroprotective profile, which consisted of lower total and LDL cholesterol levels. Plasma from chronic ICH-CAA patients also showed a redistribution of ApoC-III from HDL to VLDL and a higher ApoE/ApoC-III ratio in HDL. Whether these alterations reflect a protective response or have a causative effect on the pathology requires further investigation.
Collapse
|
17
|
Koike T, Koike Y, Yang D, Guo Y, Rom O, Song J, Xu J, Chen Y, Wang Y, Zhu T, Garcia-Barrio MT, Fan J, Chen YE, Zhang J. Human apolipoprotein A-II reduces atherosclerosis in knock-in rabbits. Atherosclerosis 2021; 316:32-40. [PMID: 33296791 PMCID: PMC7770079 DOI: 10.1016/j.atherosclerosis.2020.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Apolipoprotein A-II (apoAII) is the second major apolipoprotein of the high-density lipoprotein (HDL) particle, after apoAI. Unlike apoAI, the biological and physiological functions of apoAII are unclear. We aimed to gain insight into the specific roles of apoAII in lipoprotein metabolism and atherosclerosis using a novel rabbit model. METHODS Wild-type (WT) rabbits are naturally deficient in apoAII, thus their HDL contains only apoAI. Using TALEN technology, we replaced the endogenous apoAI in rabbits through knock-in (KI) of human apoAII. The newly generated apoAII KI rabbits were used to study the specific function of apoAII, independent of apoAI. RESULTS ApoAII KI rabbits expressed exclusively apoAII without apoAI, as confirmed by RT-PCR and Western blotting. On a standard diet, the KI rabbits exhibited lower plasma triglycerides (TG, 52%, p < 0.01) due to accelerated clearance of TG-rich particles and higher lipoprotein lipase activity than the WT littermates. ApoAII KI rabbits also had higher plasma HDL-C (28%, p < 0.05) and their HDL was rich in apoE, apoAIV, and apoAV. When fed a cholesterol-rich diet for 16 weeks, apoAII KI rabbits were resistant to diet-induced hypertriglyceridemia and developed significantly less aortic atherosclerosis compared to WT rabbits. HDL isolated from rabbits with apoAII KI had similar cholesterol efflux capacity and anti-inflammatory effects as HDL isolated from the WT rabbits. CONCLUSIONS ApoAII KI rabbits developed less atherosclerosis than WT rabbits, possibly through increased plasma HDL-C, reduced TG and atherogenic lipoproteins. These results suggest that apoAII may serve as a potential target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Tomonari Koike
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA; Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Yamanashi, Japan
| | - Yui Koike
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yanhong Guo
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Oren Rom
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jun Song
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yajie Chen
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Yamanashi, Japan
| | - Yanli Wang
- Department of Pathology, Xi'an Medical University, Xi'an, China
| | - Tianqing Zhu
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Minerva T Garcia-Barrio
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Yamanashi, Japan.
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Chew H, Solomon VA, Fonteh AN. Involvement of Lipids in Alzheimer's Disease Pathology and Potential Therapies. Front Physiol 2020; 11:598. [PMID: 32581851 PMCID: PMC7296164 DOI: 10.3389/fphys.2020.00598] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Lipids constitute the bulk of the dry mass of the brain and have been associated with healthy function as well as the most common pathological conditions of the brain. Demographic factors, genetics, and lifestyles are the major factors that influence lipid metabolism and are also the key components of lipid disruption in Alzheimer's disease (AD). Additionally, the most common genetic risk factor of AD, APOE ϵ4 genotype, is involved in lipid transport and metabolism. We propose that lipids are at the center of Alzheimer's disease pathology based on their involvement in the blood-brain barrier function, amyloid precursor protein (APP) processing, myelination, membrane remodeling, receptor signaling, inflammation, oxidation, and energy balance. Under healthy conditions, lipid homeostasis bestows a balanced cellular environment that enables the proper functioning of brain cells. However, under pathological conditions, dyshomeostasis of brain lipid composition can result in disturbed BBB, abnormal processing of APP, dysfunction in endocytosis/exocytosis/autophagocytosis, altered myelination, disturbed signaling, unbalanced energy metabolism, and enhanced inflammation. These lipid disturbances may contribute to abnormalities in brain function that are the hallmark of AD. The wide variance of lipid disturbances associated with brain function suggest that AD pathology may present as a complex interaction between several metabolic pathways that are augmented by risk factors such as age, genetics, and lifestyles. Herewith, we examine factors that influence brain lipid composition, review the association of lipids with all known facets of AD pathology, and offer pointers for potential therapies that target lipid pathways.
Collapse
Affiliation(s)
- Hannah Chew
- Huntington Medical Research Institutes, Pasadena, CA, United States
- University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Alfred N. Fonteh
- Huntington Medical Research Institutes, Pasadena, CA, United States
| |
Collapse
|
19
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
20
|
Sabherwal S, Föcking M, English JA, Fitzsimons S, Hryniewiecka M, Wynne K, Scaife C, Healy C, Cannon M, Belton O, Zammit S, Cagney G, Cotter DR. ApoE elevation is associated with the persistence of psychotic experiences from age 12 to age 18: Evidence from the ALSPAC birth cohort. Schizophr Res 2019; 209:141-147. [PMID: 31080155 DOI: 10.1016/j.schres.2019.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/04/2019] [Accepted: 05/01/2019] [Indexed: 11/24/2022]
Abstract
Apolipoproteins, which play important roles in lipid metabolism, innate immunity and synaptic signalling, have been implicated in first episode psychosis and schizophrenia. This is the first study to investigate plasma apolipoprotein expression in children with psychotic experiences that persist into adulthood. Here, using semi-targeted proteomic analysis we compared plasma apolipoprotein expression levels in age 12 subjects who reported psychotic experiences at both age 12 and age 18 (n = 37) with age-matched subjects who only experienced psychotic experiences (PEs) at age 12 (n = 38). Participants were recruited from the UK Avon Longitudinal Study of Parents and Children (ALSPAC) cohort who participated in psychiatric assessment interviews at ages 12 and 18. We identified apoE, a protein with significant regulatory activity on cholesterol metabolism in the brain, to be significantly up regulated (p < 0.003) in those with persistent psychotic experiences. We confirmed this finding in these samples using ELISA. Our findings indicate elevated plasma apoE in age 12 children who experience PEs is associated with persistence psychotic experiences.
Collapse
Affiliation(s)
- Sophie Sabherwal
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland.
| | - Melanie Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland
| | - Jane A English
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland
| | - Stephen Fitzsimons
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Magdalena Hryniewiecka
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland
| | - Kieran Wynne
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Caitriona Scaife
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Colm Healy
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland
| | - Orina Belton
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Stanley Zammit
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK; Centre for Academic Mental Health, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gerard Cagney
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - David R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland (RCSI), Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
21
|
Henrich SE, Thaxton CS. An update on synthetic high-density lipoprotein-like nanoparticles for cancer therapy. Expert Rev Anticancer Ther 2019; 19:515-528. [DOI: 10.1080/14737140.2019.1624529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Stephen E. Henrich
- Department of Urology, Simpson Querrey Institute for BioNanotechnology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - C. Shad Thaxton
- Department of Urology, Simpson Querrey Institute for BioNanotechnology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
22
|
Wagner R, Dittrich J, Thiery J, Ceglarek U, Burkhardt R. Simultaneous LC/MS/MS quantification of eight apolipoproteins in normal and hypercholesterolemic mouse plasma. J Lipid Res 2019; 60:900-908. [PMID: 30723096 PMCID: PMC6446716 DOI: 10.1194/jlr.d084301] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
Apolipoproteins are major structural and functional constituents of lipoprotein particles. As modulators of lipid metabolism, adipose tissue biology, and energy homeostasis, apolipoproteins may serve as biomarkers or potential therapeutic targets for cardiometabolic diseases. Mice are the preferred model to study metabolic disease and CVD, but a comprehensive method to quantify circulating apolipoproteins in mice is lacking. We developed and validated a targeted proteomics assay to quantify eight apolipoproteins in mice via proteotypic signature peptides and corresponding stable isotope-labeled analogs. The LC/MS/MS method requires only a 3 µl sample volume to simultaneously determine mouse apoA-I, apoA-II, apoA-IV, apoB-100, total apoB, apoC-I, apoE, and apoJ concentrations. ApoB-48 concentrations can be calculated by subtracting apoB-100 from total apoB. After we established the analytic performance (sensitivity, linearity, and imprecision) and compared results for selected apolipoproteins against immunoassays, we applied the method to profile apolipoprotein levels in plasma and isolated HDL from normocholesterolemic C57BL/6 mice and from hypercholesterolemic Ldl-receptor- and Apoe-deficient mice. In conclusion, we present a robust, quantitative LC/MS/MS method for the multiplexed analysis of eight apolipoproteins in mice. This assay can be applied to investigate the effects of genetic manipulation or dietary interventions on apolipoprotein levels in plasma and isolated lipoprotein fractions.
Collapse
Affiliation(s)
- Richard Wagner
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Julia Dittrich
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Germany.
| | - Ralph Burkhardt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
23
|
Lee SE, Schulze K, Stewart CP, Cole RN, Wu LSF, Eroglu A, Yager JD, Groopman J, Christian P, West KP. Plasma proteome correlates of lipid and lipoprotein: biomarkers of metabolic diversity and inflammation in children of rural Nepal. J Lipid Res 2018; 60:149-160. [PMID: 30473544 PMCID: PMC6314253 DOI: 10.1194/jlr.p088542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Proteins involved in lipoprotein metabolism can modulate cardiovascular health. While often measured to assess adult metabolic diseases, little is known about the proteomes of lipoproteins and their relation to metabolic dysregulation and underlying inflammation in undernourished child populations. The objective of this population study was to globally characterize plasma proteins systemically associated with HDL, LDL, and triglycerides in 500 Nepalese children. Abnormal lipid profiles characterized by elevated plasma triglycerides and low HDL-cholesterol (HDL-C) concentrations were common, especially in children with subclinical inflammation. Among 982 proteins analyzed, the relative abundance of 11, 12, and 52 plasma proteins was correlated with LDL-cholesterol (r = −0.43∼0.70), triglycerides (r = −0.39∼0.53), and HDL-C (r = −0.49∼0.79) concentrations, respectively. These proteins included apolipoproteins and numerous unexpected intracellular and extracellular matrix binding proteins, likely originating in hepatic and peripheral tissues. Relative abundance of two-thirds of the HDL proteome varied with inflammation, with acute phase reactants higher by 4∼40%, and proteins involved in HDL biosynthesis, cholesterol efflux, vitamin transport, angiogenesis, and tissue repair lower by 3∼20%. Untargeted plasma proteomics detects comprehensive sets of both known and novel lipoprotein-associated proteins likely reflecting systemic regulation of lipoprotein metabolism and vascular homeostasis. Inflammation-altered distributions of the HDL proteome may be predisposing undernourished populations to early chronic disease.
Collapse
Affiliation(s)
- Sun Eun Lee
- Center for Human Nutrition Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kerry Schulze
- Center for Human Nutrition Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Christine P Stewart
- Program in International and Community Nutrition, Department of Nutrition, University of California, Davis, CA
| | - Robert N Cole
- Mass Spectrometry and Proteomics Facility, Johns Hopkins School of Medicine, Baltimore, MD
| | - Lee S-F Wu
- Center for Human Nutrition Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Abdulkerim Eroglu
- Center for Human Nutrition Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - James D Yager
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - John Groopman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Parul Christian
- Center for Human Nutrition Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Keith P West
- Center for Human Nutrition Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
24
|
Mazidi M, Mikhailidis DP, Banach M. Associations between risk of overall mortality, cause-specific mortality and level of inflammatory factors with extremely low and high high-density lipoprotein cholesterol levels among American adults. Int J Cardiol 2018; 276:242-247. [PMID: 30473336 DOI: 10.1016/j.ijcard.2018.11.095] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/12/2018] [Accepted: 11/15/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND The health outcomes associated with extremely low or high plasma concentrations of high-density lipoprotein cholesterol (HDL-C) are not well documented mainly because of the small numbers of participants with such values included in the clinical trials. OBJECTIVE We prospectively investigated the association between extremely low and high HDL-C with: 1) the risk of overall, coronary heart disease (CHD), cerebrovascular and cancer mortality, and, 2) their link with inflammatory factors. METHODS Analysis was based on subjects ≥18 years old from the National Health and Nutrition Examination Surveys (NHANES). We categorized HDL-C levels as follows: [low HDL-C group ≤30 (extremely low), 30-40 (low), and ≥40 (reference)] [high HDL-C group = 40-80 (reference), 80-100 (high) and ≥100 mg/dl (extremely high). Cox proportional hazard regression models and analysis of covariance accounted for survey design, masked variance and sample weights. RESULTS After adjustment for age, race and sex, we found that the very low HDL-C category (<30 mg/dl) had a greater risk of total mortality (risk ratio [RR]: 3.00, 95%CI: 2.20-4.09). RR for CHD and stroke mortality was 2.00 and 2.53, respectively; there was no link between cancer and level of HDL-C (p = 0.235). The association between total mortality, CHD and stroke with the level of HDL-C attenuated but remained significant even after adjustment for demographics, dietary, cardiovascular risk factors and treatment for dyslipidemia (all p < 0.001). After adjustments, subjects with extremely high HDL-C levels had a higher risk of mortalities (all p < 0.001). Mexican-American ethnicity, subjects in the low level of HDL-C (30-40 mg/dl) category had higher risk of mortalities than those with a very low level (all p < 0.001). Concentration of C-reactive protein, fibrinogen and white blood count significantly decreased as the level of the HDL-C increased; these findings were robust after adjustment for demographics, dietary, cardiovascular risk factors and treatment for dyslipidemia (all p < 0.001); further subjects with extremely high HDL-C levels have a greater levels of inflammatory factors (all p < 0.001). CONCLUSIONS Both extremely low and high HDL-C levels were associated with greater risk of mortalities (total, CHD and stroke) and higher level of inflammatory factors, while there was no link between level of HDL-C and risk of cancer. Moreover, we found evidence of an HDL-C paradox in Mexican-American ethnicity participants.
Collapse
Affiliation(s)
- Mohsen Mazidi
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, UK
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland.
| |
Collapse
|
25
|
Vezočnik V, Hodnik V, Sitar S, Okur HI, Tušek-Žnidarič M, Lütgebaucks C, Sepčić K, Kogej K, Roke S, Žagar E, Maček P. Kinetically Stable Triglyceride-Based Nanodroplets and Their Interactions with Lipid-Specific Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:8983-8993. [PMID: 29983071 DOI: 10.1021/acs.langmuir.8b02180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Understanding of the interactions between proteins and natural and artificially prepared lipid membrane surfaces and embedded nonpolar cores is important in studies of physiological processes and their pathologies and is applicable to nanotechnologies. In particular, rapidly growing interest in cellular droplets defines the need for simplified biomimetic lipid model systems to overcome in vivo complexity and variability. We present a protocol for the preparation of kinetically stable nanoemulsions with nanodroplets composed of sphingomyelin (SM) and cholesterol (Chol), as amphiphilic surfactants, and trioleoylglycerol (TOG), at various molar ratios. To prepare stable SM/Chol-coated monodisperse lipid nanodroplets, we modified a reverse phase evaporation method and combined it with ultrasonication. Lipid composition, ζ-potential, gyration and hydrodynamic radius, shape, and temporal stability of the lipid nanodroplets were characterized and compared to extruded SM/Chol large unilamellar vesicles. Lipid nanodroplets and large unilamellar vesicles with theoretical SM/Chol/TOG molar ratios of 1/1/4.7 and 4/1/11.7 were further investigated for the orientational order of their interfacial water molecules using a second harmonic scattering technique, and for interactions with the SM-binding and Chol-binding pore-forming toxins equinatoxin II and perfringolysin O, respectively. The surface characteristics (ζ-potential, orientational order of interfacial water molecules) and binding of these proteins to the nanodroplet SM/Chol monolayers were similar to those for the SM/Chol bilayers of the large unilamellar vesicles and SM/Chol Langmuir monolayers, in terms of their surface structures. We propose that such SM/Chol/TOG nanoparticles with the required lipid compositions can serve as experimental models for monolayer membrane to provide a system that imitates the natural lipid droplets.
Collapse
Affiliation(s)
- Valerija Vezočnik
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| | - Simona Sitar
- Department of Polymer Chemistry and Technology , National Institute of Chemistry , Hajdrihova 19 , Ljubljana 1000 , Slovenia
| | - Halil I Okur
- Laboratory for Fundamental BioPhotonics, Institute of Bio-Engineering, and Institute of Material Science, School of Engineering, and Lausanne Centre for Ultrafast Science , École Polytechnique Fédérale de Lausanne , CH-1015 Lausanne , Switzerland
| | | | - Cornelis Lütgebaucks
- Laboratory for Fundamental BioPhotonics, Institute of Bio-Engineering, and Institute of Material Science, School of Engineering, and Lausanne Centre for Ultrafast Science , École Polytechnique Fédérale de Lausanne , CH-1015 Lausanne , Switzerland
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| | - Ksenija Kogej
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology , University of Ljubljana , Večna pot 113 , Ljubljana 1000 , Slovenia
| | - Sylvie Roke
- Laboratory for Fundamental BioPhotonics, Institute of Bio-Engineering, and Institute of Material Science, School of Engineering, and Lausanne Centre for Ultrafast Science , École Polytechnique Fédérale de Lausanne , CH-1015 Lausanne , Switzerland
| | - Ema Žagar
- Department of Polymer Chemistry and Technology , National Institute of Chemistry , Hajdrihova 19 , Ljubljana 1000 , Slovenia
| | - Peter Maček
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| |
Collapse
|
26
|
Liang K, Gao H, Gu Y, Yang S, Zhang J, Li J, Wang Y, Wang Y, Li Y. Graphene oxide aggregate-assisted LDI-MS for the direct analysis of triacylglycerol in complex biological samples. Anal Chim Acta 2018; 1035:108-118. [PMID: 30224128 DOI: 10.1016/j.aca.2018.07.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/18/2018] [Accepted: 07/22/2018] [Indexed: 12/13/2022]
Abstract
Knowledge of blood triacylglycerol (TAG) species is essential to clarify the physiological functions of individual TAG molecules and also to develop potential biomarkers for related diseases. Commonly, lipid samples prepared by organic liquid-liquid extraction contain complex components, thus cannot be directly characterized by mass spectrometry (MS) and often require an additional purification step. Here, we described a laser desorption ionization - mass spectrometry (LDI-MS) method that utilized aggregated graphene oxide (AGO) as both lipid extractant and MS matrix (AGOLDI-MS), to characterize and quantify plasma TAG species without the use of harmful solvent or complex separation step. We first designed and synthesized the AGO material with a multi-layered sheet structure, which could efficiently break up the structure of lipoproteins, and extract plasma TAGs as solid-phase extraction material. Furthermore, in AGOLDI-MS procedure, the AGO could directly act as matrix and selectively produce the MS signals of TAGs without the interferences of phospholipids, which was hardly achieved by using the routine LDI-MS method based on liquid-liquid extraction and small molecular matrix. We confirmed the suitability of AGOLDI-MS as characterization and quantitative tool for TAG species through studying the analysis performances in TAG standards and real plasma samples. To establish potential utility of our method, we characterized 42 human plasmas from healthy and hyperlipemic donators, indicating that the AGOLDI-MS could not only generate comparable quantitative results of total TAGs to current clinical technology, but also monitor the changes of TAG species between different sample groups. This approach could further characterize the compositions of the fatty acid moieties in even low abundant TAGs by the assistance of tandem MS-MS. This concise, specific, and high-throughput approach will facilitate the rapid and precise characterizations of plasma TAGs, and make the MS approach for TAGs more adaptable for clinical uses.
Collapse
Affiliation(s)
- Kai Liang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Huanyu Gao
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yajun Gu
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Shaojie Yang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; GuangDong Bio-healtech Advanced Co., Ltd, Foshan City, GuangDong Province, 52800, China
| | | | - Jiejie Li
- Beijing Tian Tan Hospital, Beijing, 100050, China
| | - Yilong Wang
- Beijing Tian Tan Hospital, Beijing, 100050, China
| | - Yongjun Wang
- Beijing Tian Tan Hospital, Beijing, 100050, China
| | - Yan Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
27
|
Lutomski CA, Gordon SM, Remaley AT, Jarrold MF. Resolution of Lipoprotein Subclasses by Charge Detection Mass Spectrometry. Anal Chem 2018; 90:6353-6356. [DOI: 10.1021/acs.analchem.8b01127] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Corinne A. Lutomski
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Scott M. Gordon
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Martin F. Jarrold
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
28
|
Kuklenyik Z, Jones JI, Gardner MS, Schieltz DM, Parks BA, Toth CA, Rees JC, Andrews ML, Carter K, Lehtikoski AK, McWilliams LG, Williamson YM, Bierbaum KP, Pirkle JL, Barr JR. Core lipid, surface lipid and apolipoprotein composition analysis of lipoprotein particles as a function of particle size in one workflow integrating asymmetric flow field-flow fractionation and liquid chromatography-tandem mass spectrometry. PLoS One 2018; 13:e0194797. [PMID: 29634782 PMCID: PMC5892890 DOI: 10.1371/journal.pone.0194797] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/09/2018] [Indexed: 12/18/2022] Open
Abstract
Lipoproteins are complex molecular assemblies that are key participants in the intricate cascade of extracellular lipid metabolism with important consequences in the formation of atherosclerotic lesions and the development of cardiovascular disease. Multiplexed mass spectrometry (MS) techniques have substantially improved the ability to characterize the composition of lipoproteins. However, these advanced MS techniques are limited by traditional pre-analytical fractionation techniques that compromise the structural integrity of lipoprotein particles during separation from serum or plasma. In this work, we applied a highly effective and gentle hydrodynamic size based fractionation technique, asymmetric flow field-flow fractionation (AF4), and integrated it into a comprehensive tandem mass spectrometry based workflow that was used for the measurement of apolipoproteins (apos A-I, A-II, A-IV, B, C-I, C-II, C-III and E), free cholesterol (FC), cholesterol esters (CE), triglycerides (TG), and phospholipids (PL) (phosphatidylcholine (PC), sphingomyelin (SM), phosphatidylethanolamine (PE), phosphatidylinositol (PI) and lysophosphatidylcholine (LPC)). Hydrodynamic size in each of 40 size fractions separated by AF4 was measured by dynamic light scattering. Measuring all major lipids and apolipoproteins in each size fraction and in the whole serum, using total of 0.1 ml, allowed the volumetric calculation of lipoprotein particle numbers and expression of composition in molar analyte per particle number ratios. Measurements in 110 serum samples showed substantive differences between size fractions of HDL and LDL. Lipoprotein composition within size fractions was expressed in molar ratios of analytes (A-I/A-II, C-II/C-I, C-II/C-III. E/C-III, FC/PL, SM/PL, PE/PL, and PI/PL), showing differences in sample categories with combinations of normal and high levels of Total-C and/or Total-TG. The agreement with previous studies indirectly validates the AF4-LC-MS/MS approach and demonstrates the potential of this workflow for characterization of lipoprotein composition in clinical studies using small volumes of archived frozen samples.
Collapse
Affiliation(s)
- Zsuzsanna Kuklenyik
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jeffery I. Jones
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Michael S. Gardner
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - David M. Schieltz
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Bryan A. Parks
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Christopher A. Toth
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jon C. Rees
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Michael L. Andrews
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Kayla Carter
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Antony K. Lehtikoski
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lisa G. McWilliams
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Yulanda M. Williamson
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Kevin P. Bierbaum
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James L. Pirkle
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - John R. Barr
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
29
|
Christinat N, Masoodi M. Comprehensive Lipoprotein Characterization Using Lipidomics Analysis of Human Plasma. J Proteome Res 2017. [DOI: 10.1021/acs.jproteome.7b00236] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Nicolas Christinat
- Lipid Biology, Nestlé Institute of Health Sciences, EPFL Innovation Park, Bâtiment
H, 1015 Lausanne, Switzerland
| | - Mojgan Masoodi
- Lipid Biology, Nestlé Institute of Health Sciences, EPFL Innovation Park, Bâtiment
H, 1015 Lausanne, Switzerland
| |
Collapse
|
30
|
Kaul S, Xu H, Zabalawi M, Maruko E, Fulp BE, Bluemn T, Brzoza-Lewis KL, Gerelus M, Weerasekera R, Kallinger R, James R, Zhang YS, Thomas MJ, Sorci-Thomas MG. Lipid-Free Apolipoprotein A-I Reduces Progression of Atherosclerosis by Mobilizing Microdomain Cholesterol and Attenuating the Number of CD131 Expressing Cells: Monitoring Cholesterol Homeostasis Using the Cellular Ester to Total Cholesterol Ratio. J Am Heart Assoc 2016; 5:JAHA.116.004401. [PMID: 27821400 PMCID: PMC5210328 DOI: 10.1161/jaha.116.004401] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disorder whose development is inversely correlated with high-density lipoprotein concentration. Current therapies involve pharmaceuticals that significantly elevate plasma high-density lipoprotein cholesterol concentrations. Our studies were conducted to investigate the effects of low-dose lipid-free apolipoprotein A-I (apoA-I) on chronic inflammation. The aims of these studies were to determine how subcutaneously injected lipid-free apoA-I reduces accumulation of lipid and immune cells within the aortic root of hypercholesterolemic mice without sustained elevations in plasma high-density lipoprotein cholesterol concentrations. METHODS AND RESULTS Ldlr-/- and Ldlr-/- apoA-I-/- mice were fed a Western diet for a total of 12 weeks. After 6 weeks, a subset of mice from each group received subcutaneous injections of 200 μg of lipid-free human apoA-I 3 times a week, while the other subset received 200 μg of albumin, as a control. Mice treated with lipid-free apoA-I showed a decrease in cholesterol deposition and immune cell retention in the aortic root compared with albumin-treated mice, regardless of genotype. This reduction in atherosclerosis appeared to be directly related to a decrease in the number of CD131 expressing cells and the esterified cholesterol to total cholesterol content in several immune cell compartments. In addition, apoA-I treatment altered microdomain cholesterol composition that shifted CD131, the common β subunit of the interleukin 3 receptor, from lipid raft to nonraft fractions of the plasma membrane. CONCLUSIONS ApoA-I treatment reduced lipid and immune cell accumulation within the aortic root by systemically reducing microdomain cholesterol content in immune cells. These data suggest that lipid-free apoA-I mediates beneficial effects through attenuation of immune cell lipid raft cholesterol content, which affects numerous types of signal transduction pathways that rely on microdomain integrity for assembly and activation.
Collapse
Affiliation(s)
- Sushma Kaul
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Hao Xu
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Manal Zabalawi
- Section of Molecular Medicine, and Biochemistry, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Elisa Maruko
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Brian E Fulp
- Section of Molecular Medicine, and Biochemistry, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Theresa Bluemn
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Kristina L Brzoza-Lewis
- Section of Molecular Medicine, and Biochemistry, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Mark Gerelus
- Section of Molecular Medicine, and Biochemistry, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Rachel Kallinger
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Roland James
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.,Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI.,TOPS Obesity and Metabolic Research Center, Medical College of Wisconsin, Milwaukee, WI
| | - Yi Sherry Zhang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.,Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI.,TOPS Obesity and Metabolic Research Center, Medical College of Wisconsin, Milwaukee, WI
| | - Michael J Thomas
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Mary G Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI .,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
31
|
Tada H, Kawashiri MA, Konno T, Nohara A, Inazu A, Mabuchi H, Yamagishi M, Hayashi K. Prevalence, clinical features, and prognosis of patients with extremely low high-density lipoprotein cholesterol. J Clin Lipidol 2016; 10:1311-1317. [DOI: 10.1016/j.jacl.2016.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 06/28/2016] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
|
32
|
Henderson CM, Vaisar T, Hoofnagle AN. Isolating and Quantifying Plasma HDL Proteins by Sequential Density Gradient Ultracentrifugation and Targeted Proteomics. Methods Mol Biol 2016; 1410:105-20. [PMID: 26867741 PMCID: PMC5501989 DOI: 10.1007/978-1-4939-3524-6_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The sensitivity and specificity of tandem mass spectrometers have made targeted proteomics the method of choice for the precise simultaneous measurement of many proteins in complex mixtures. Its application to the relative quantification of proteins in high-density lipoproteins (HDL) that have been purified from human plasma has revealed potential mechanisms to explain the atheroprotective effects of HDL. We describe a moderate throughput method for isolating HDL from human plasma that uses sequential density gradient ultracentrifugation, the traditional method of HDL purification, and subsequent trypsin digestion and nanoflow liquid chromatography-tandem mass spectrometry to quantify 38 proteins in the HDL fraction of human plasma. To control for the variability associated with digestion, matrix effects, and instrument performance, we normalize the signal from endogenous HDL protein-associated peptides liberated during trypsin digestion to the signal from peptides liberated from stable isotope-labeled apolipoprotein A-I spiked in as an internal standard prior to digestion. The method has good reproducibility and other desirable characteristics for preclinical research.
Collapse
Affiliation(s)
- Clark M Henderson
- Department of Laboratory Medicine, University of Washington School of Medicine, Box 357110, Seattle, WA, 98195-7110, USA
| | - Tomas Vaisar
- Department of Medicine, University of Washington School of Medicine, Box 358055, Seattle, WA, 98195-8055, USA
| | - Andrew N Hoofnagle
- Department of Laboratory Medicine, University of Washington School of Medicine, Box 357110, Seattle, WA, 98195-7110, USA.
- Department of Medicine, University of Washington School of Medicine, Box 358055, Seattle, WA, 98195-8055, USA.
| |
Collapse
|
33
|
Singh SA, Miyosawa K, Aikawa M. Mass spectrometry meets the challenge of understanding the complexity of the lipoproteome: recent findings regarding proteins involved in dyslipidemia and cardiovascular disease. Expert Rev Proteomics 2015; 12:519-32. [PMID: 26325144 DOI: 10.1586/14789450.2015.1078731] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite the fact that link between dyslipidemia and atherosclerosis was made over 100 years ago, atherosclerosis remains a major cause of morbidity and mortality worldwide. Major efforts focus towards understanding lipid metabolism, particularly by studying its particle compartments in circulation: the lipoproteins. In recent years, mass spectrometry has played an integral role in the deep sequencing of the lipoproteome and in metabolism studies conducted in vivo. This review highlights the path of lipoprotein research towards state-of-the-art mass spectrometry with special emphasis on the method of selected reaction monitoring and its impact on apolipoprotein metabolism studies. Also presented is what is expected for the lipoprotein field with the recent advent of high resolution/accurate mass parallel reaction monitoring mass spectrometry. The benefits of high resolution/accurate mass measurements are demonstrated by example instrument workflows and by detailing a novel method to quantify very low levels of circulating proprotein convertase subtilisin-kexin type 9 in rabbit. It is anticipated that future clinical studies or clinical trials aimed to treat dyslipidemia by manipulating key regulatory proteins will benefit from the new and exciting opportunities afforded by the latest technologies in mass spectrometry.
Collapse
Affiliation(s)
- Sasha A Singh
- a 1 Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Katsutoshi Miyosawa
- a 1 Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- a 1 Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,b 2 Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
34
|
Dissecting the proteome of lipoproteins: New biomarkers for cardiovascular diseases? TRANSLATIONAL PROTEOMICS 2015. [DOI: 10.1016/j.trprot.2014.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
35
|
Vaisar T, Tang C, Babenko I, Hutchins P, Wimberger J, Suffredini AF, Heinecke JW. Inflammatory remodeling of the HDL proteome impairs cholesterol efflux capacity. J Lipid Res 2015; 56:1519-30. [PMID: 25995210 DOI: 10.1194/jlr.m059089] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Indexed: 12/17/2022] Open
Abstract
Recent studies demonstrate that HDL's ability to promote cholesterol efflux from macrophages associates strongly with cardioprotection in humans independently of HDL-cholesterol (HDL-C) and apoA-I, HDL's major protein. However, the mechanisms that impair cholesterol efflux capacity during vascular disease are unclear. Inflammation, a well-established risk factor for cardiovascular disease, has been shown to impair HDL's cholesterol efflux capacity. We therefore tested the hypothesis that HDL's impaired efflux capacity is mediated by specific changes of its protein cargo. Humans with acute inflammation induced by low-level endotoxin had unchanged HDL-C levels, but their HDL-C efflux capacity was significantly impaired. Proteomic analyses demonstrated that HDL's cholesterol efflux capacity correlated inversely with HDL content of serum amyloid A (SAA)1 and SAA2. In mice, acute inflammation caused a marked impairment of HDL-C efflux capacity that correlated with a large increase in HDL SAA. In striking contrast, the efflux capacity of mouse inflammatory HDL was preserved with genetic ablation of SAA1 and SAA2. Our observations indicate that the inflammatory impairment of HDL-C efflux capacity is due in part to SAA-mediated remodeling of HDL's protein cargo.
Collapse
Affiliation(s)
- Tomáš Vaisar
- Department of Medicine, University of Washington, Seattle, WA 98105
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle, WA 98105
| | - Ilona Babenko
- Department of Medicine, University of Washington, Seattle, WA 98105
| | - Patrick Hutchins
- Department of Medicine, University of Washington, Seattle, WA 98105
| | - Jake Wimberger
- Department of Medicine, University of Washington, Seattle, WA 98105
| | - Anthony F Suffredini
- Critical Care Medicine Department, National Institutes of Health, Bethesda, MD 20892
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, WA 98105
| |
Collapse
|
36
|
Rached FH, Chapman MJ, Kontush A. HDL particle subpopulations: Focus on biological function. Biofactors 2015; 41:67-77. [PMID: 25809447 DOI: 10.1002/biof.1202] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/04/2015] [Accepted: 02/07/2015] [Indexed: 12/12/2022]
Abstract
Low levels of high-density lipoprotein-cholesterol (HDL-C) constitute an independent biomarker of cardiovascular morbi-mortality. However, recent advances have drastically modified the classical and limited view of HDL as a carrier of 'good cholesterol', and have revealed unexpected levels of complexity in the circulating HDL particle pool. HDL particles are indeed highly heterogeneous in structure, intravascular metabolism and biological activity. This review describes recent progress in our understanding of HDL subpopulations and their biological activities, and focuses on relationships between the structural, compositional and functional heterogeneity of HDL particles.
Collapse
Affiliation(s)
- Fabiana H Rached
- National Institute for Health and Medical Research (INSERM), UMR-ICAN 1166, Université Pierre et Marie Curie-Paris 6, AP-HP, Pitié-Salpétrière University Hospital, ICAN, Paris, France; Heart Institute-InCor, University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | | |
Collapse
|
37
|
Mangrum JB, Martin EJ, Brophy DF, Hawkridge AM. Intact stable isotope labeled plasma proteins from the SILAC-labeled HepG2 secretome. Proteomics 2015; 15:3104-15. [DOI: 10.1002/pmic.201400369] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/05/2014] [Accepted: 12/05/2014] [Indexed: 01/26/2023]
Affiliation(s)
- John B. Mangrum
- Department of Pharmaceutics; Virginia Commonwealth University; Richmond VA USA
| | - Erika J. Martin
- Department of Pharmacotherapy & Outcomes Science; Virginia Commonwealth University; Richmond VA USA
| | - Donald F. Brophy
- Department of Pharmacotherapy & Outcomes Science; Virginia Commonwealth University; Richmond VA USA
| | - Adam M. Hawkridge
- Department of Pharmaceutics; Virginia Commonwealth University; Richmond VA USA
- Department of Pharmacotherapy & Outcomes Science; Virginia Commonwealth University; Richmond VA USA
| |
Collapse
|
38
|
Kontush A, Lindahl M, Lhomme M, Calabresi L, Chapman MJ, Davidson WS. Structure of HDL: particle subclasses and molecular components. Handb Exp Pharmacol 2015; 224:3-51. [PMID: 25522985 DOI: 10.1007/978-3-319-09665-0_1] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A molecular understanding of high-density lipoprotein (HDL) will allow a more complete grasp of its interactions with key plasma remodelling factors and with cell-surface proteins that mediate HDL assembly and clearance. However, these particles are notoriously heterogeneous in terms of almost every physical, chemical and biological property. Furthermore, HDL particles have not lent themselves to high-resolution structural study through mainstream techniques like nuclear magnetic resonance and X-ray crystallography; investigators have therefore had to use a series of lower resolution methods to derive a general structural understanding of these enigmatic particles. This chapter reviews current knowledge of the composition, structure and heterogeneity of human plasma HDL. The multifaceted composition of the HDL proteome, the multiple major protein isoforms involving translational and posttranslational modifications, the rapidly expanding knowledge of the HDL lipidome, the highly complex world of HDL subclasses and putative models of HDL particle structure are extensively discussed. A brief history of structural studies of both plasma-derived and recombinant forms of HDL is presented with a focus on detailed structural models that have been derived from a range of techniques spanning mass spectrometry to molecular dynamics.
Collapse
Affiliation(s)
- Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMR-ICAN 1166, Paris, France,
| | | | | | | | | | | |
Collapse
|
39
|
Kratzer A, Giral H, Landmesser U. High-density lipoproteins as modulators of endothelial cell functions: alterations in patients with coronary artery disease. Cardiovasc Res 2014; 103:350-61. [DOI: 10.1093/cvr/cvu139] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
40
|
von Zychlinski A, Williams M, McCormick S, Kleffmann T. Absolute quantification of apolipoproteins and associated proteins on human plasma lipoproteins. J Proteomics 2014; 106:181-90. [DOI: 10.1016/j.jprot.2014.04.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/14/2014] [Accepted: 04/18/2014] [Indexed: 10/25/2022]
|
41
|
Gugliucci A, Caccavello R, Kotani K, Kimura S. Evidence for the presence of active paraoxonase 1 in small-dense low-density lipoprotein. Redox Rep 2014; 19:154-60. [PMID: 24524596 DOI: 10.1179/1351000214y.0000000084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
AIM We have recently demonstrated the quick ex vivo transfer of paraoxonase 1 (PON1) activity from high-density lipoprotein (HDL) to small, dense low-density lipoprotein (sdLDL). We set out to assess whether sdLDL contains active PON1 in vivo. METHODS We conducted a nested case-control, proof of principle study with the Japanese healthy subjects with normal lipids (n = 23) and age and gender-paired dyslipidemic subjects (n = 17). Lipid panels, lactonase and arylesterase assays, and PON1 zymogram in the LDL and HDL subclasses were assessed. RESULTS PON1 specific activity in the high-molecular weight lipoprotein fraction corresponding to LDL migration was found in 48% of normo and in 29% of dyslipidemic Japanese subjects. This band co-localizes with apoB100 and not Lp(a) and displays a lower molecular mass than the bulk of LDL. CONCLUSION We provide evidence, for the first time, that native sdLDL contains up to 4% of the total PON1 activity in the serum of up to 48% of the Japanese subjects. Could the PON1-containing sdLDL represent a set of particles with a defense mechanism from oxidation and therefore its levels actually prove to be atheroprotective? If further studies confirm this contention, a zymogram of PON1 in LDL subclasses could be a functional assay that complements the current methods that only inform on the size and lipid concentration of these particles.
Collapse
|
42
|
Activation of paraoxonase 1 is associated with HDL remodeling ex vivo. Clin Chim Acta 2013; 429:38-45. [PMID: 24280342 DOI: 10.1016/j.cca.2013.11.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/31/2013] [Accepted: 11/16/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND We hypothesize that during high density lipoprotein (HDL) remodeling PON1 reaches an optimal distribution in HDL subclasses by which it achieves maximum activity. We conducted this study to gain insight on PON1 fate and activation during short-term HDL remodeling ex vivo. METHODS Serum from 8 healthy volunteers was either frozen at -80°C (time 0) or incubated under sterile conditions for up to 48h at 37°C or at 4°C. Aliquots were taken at 3, 6, 9, 24 and 48 h and immediately frozen at -80°C. PON1 activities were measured, as well as PON1 and apolipoprotein distributions in HDL subclasses by gradient gel electrophoresis. RESULTS The first novel finding in our study is the evidence provided for a significant activation of both lactonase and arylesterase activities of PON1 that ensues in a very short time frame of incubation of serum ex vivo at 37°C. All subjects studied displayed these changes, the activation was apparent in <3h, peaked at 6h and amounted to >20%. This is associated with a temperature and time-dependent redistribution of PON1 activity in HDL subclasses, with an increase in activity in both very large HDL2 and small HDL3 in the first phase (3-9h), followed by a progressive transfer of PON1 to very large HDL2 as the particles mature. These changes are paralleled by the appearance of weak, but apparent PON1 activity at subspecies that correspond to sdLDL. During the first phase of PON1 activation and shifts, a parallel shift of apoE can be evidenced: at 3-9h, apoE increases in sdLDL, after that time it is lost from HDL and also from sdLDL and stays in VLDL at the origin of the run. ApoA-I shifts towards larger particles, which parallels the change in PON1. As HDL matures there is a progressive shift of apoA-II towards larger HDL. Low levels of apoA-IV at the initiation of the incubation are followed by time dependent quick disappearance of apoA-IV in HDL which parallels the changes in PON1, apoE and A-II. CONCLUSION Short, ex vivo incubation of serum leads to quick activation of PON1 associated with transfers to HDL3c, large HDL and sdLDL. The process is blocked by CETP and LCAT inhibitors. The data suggest that HDL maturation optimizes PON1 activity. These findings may be of interest for future studies aimed at modulating PON-1 activity for its cardioprotective effects and suggest a new mechanism whereby CETP inhibitors failed in clinical trials.
Collapse
|
43
|
Leman LJ, Maryanoff BE, Ghadiri MR. Molecules that mimic apolipoprotein A-I: potential agents for treating atherosclerosis. J Med Chem 2013; 57:2169-96. [PMID: 24168751 DOI: 10.1021/jm4005847] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Certain amphipathic α-helical peptides can functionally mimic many of the properties of full-length apolipoproteins, thereby offering an approach to modulate high-density lipoprotein (HDL) for combating atherosclerosis. In this Perspective, we summarize the key findings and advances over the past 25 years in the development of peptides that mimic apolipoproteins, especially apolipoprotein A-I (apoA-I). This assemblage of information provides a reasonably clear picture of the state of the art in the apolipoprotein mimetic field, an appreciation of the potential for such agents in pharmacotherapy, and a sense of the opportunities for optimizing the functional properties of HDL.
Collapse
Affiliation(s)
- Luke J Leman
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | |
Collapse
|
44
|
Beaufrère H. Atherosclerosis: Comparative Pathogenesis, Lipoprotein Metabolism, and Avian and Exotic Companion Mammal Models. J Exot Pet Med 2013. [DOI: 10.1053/j.jepm.2013.10.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Adsorption of plasma proteins on uncoated PLGA nanoparticles. Eur J Pharm Biopharm 2013; 85:53-60. [DOI: 10.1016/j.ejpb.2012.11.030] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/27/2012] [Accepted: 11/28/2012] [Indexed: 01/05/2023]
|
46
|
Gordon SM, Deng J, Tomann AB, Shah AS, Lu LJ, Davidson WS. Multi-dimensional co-separation analysis reveals protein-protein interactions defining plasma lipoprotein subspecies. Mol Cell Proteomics 2013; 12:3123-34. [PMID: 23882025 DOI: 10.1074/mcp.m113.028134] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The distribution of circulating lipoprotein particles affects the risk for cardiovascular disease (CVD) in humans. Lipoproteins are historically defined by their density, with low-density lipoproteins positively and high-density lipoproteins (HDLs) negatively associated with CVD risk in large populations. However, these broad definitions tend to obscure the remarkable heterogeneity within each class. Evidence indicates that each class is composed of physically (size, density, charge) and compositionally (protein and lipid) distinct subclasses exhibiting unique functionalities and differing effects on disease. HDLs in particular contain upward of 85 proteins of widely varying function that are differentially distributed across a broad range of particle diameters. We hypothesized that the plasma lipoproteins, particularly HDL, represent a continuum of phospholipid platforms that facilitate specific protein-protein interactions. To test this idea, we separated normal human plasma using three techniques that exploit different lipoprotein physicochemical properties (gel filtration chromatography, ionic exchange chromatography, and preparative isoelectric focusing). We then tracked the co-separation of 76 lipid-associated proteins via mass spectrometry and applied a summed correlation analysis to identify protein pairs that may co-reside on individual lipoproteins. The analysis produced 2701 pairing scores, with the top hits representing previously known protein-protein interactions as well as numerous unknown pairings. A network analysis revealed clusters of proteins with related functions, particularly lipid transport and complement regulation. The specific co-separation of protein pairs or clusters suggests the existence of stable lipoprotein subspecies that may carry out distinct functions. Further characterization of the composition and function of these subspecies may point to better targeted therapeutics aimed at CVD or other diseases.
Collapse
Affiliation(s)
- Scott M Gordon
- Center for Lipid and Arteriosclerosis Science, University of Cincinnati, 2120 East Galbraith Rd., Cincinnati, Ohio 45237-0507
| | | | | | | | | | | |
Collapse
|
47
|
Chen XW, Wang H, Bajaj K, Zhang P, Meng ZX, Ma D, Bai Y, Liu HH, Adams E, Baines A, Yu G, Sartor MA, Zhang B, Yi Z, Lin J, Young SG, Schekman R, Ginsburg D. SEC24A deficiency lowers plasma cholesterol through reduced PCSK9 secretion. eLife 2013; 2:e00444. [PMID: 23580231 PMCID: PMC3622177 DOI: 10.7554/elife.00444] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 02/19/2013] [Indexed: 12/14/2022] Open
Abstract
The secretory pathway of eukaryotic cells packages cargo proteins into COPII-coated vesicles for transport from the endoplasmic reticulum (ER) to the Golgi. We now report that complete genetic deficiency for the COPII component SEC24A is compatible with normal survival and development in the mouse, despite the fundamental role of SEC24 in COPII vesicle formation and cargo recruitment. However, these animals exhibit markedly reduced plasma cholesterol, with mutations in Apoe and Ldlr epistatic to Sec24a, suggesting a receptor-mediated lipoprotein clearance mechanism. Consistent with these data, hepatic LDLR levels are up-regulated in SEC24A-deficient cells as a consequence of specific dependence of PCSK9, a negative regulator of LDLR, on SEC24A for efficient exit from the ER. Our findings also identify partial overlap in cargo selectivity between SEC24A and SEC24B, suggesting a previously unappreciated heterogeneity in the recruitment of secretory proteins to the COPII vesicles that extends to soluble as well as trans-membrane cargoes. DOI:http://dx.doi.org/10.7554/eLife.00444.001.
Collapse
Affiliation(s)
- Xiao-Wei Chen
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - He Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Kanika Bajaj
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Pengcheng Zhang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Zhuo-Xian Meng
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Danjun Ma
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, United States
| | - Yongsheng Bai
- Department of Bioinformatics, University of Michigan, Ann Arbor, United States
| | - Hui-Hui Liu
- Department of Molecular Medicine, Cleveland Clinic, Cleveland, United States
| | - Elizabeth Adams
- Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, United States
| | - Andrea Baines
- Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, United States
| | - Genggeng Yu
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Maureen A Sartor
- Department of Bioinformatics, University of Michigan, Ann Arbor, United States
| | - Bin Zhang
- Department of Molecular Medicine, Cleveland Clinic, Cleveland, United States
| | - Zhengping Yi
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, United States
| | - Jiandie Lin
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Stephen G Young
- Department of Medicine and Human Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - David Ginsburg
- Division of Molecular Medicine & Genetics, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, United States
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW To offer a comprehensive review on the role of ABCG1 in cellular sterol homeostasis. RECENT FINDINGS Early studies with Abcg1 mice indicated that ABCG1 was crucial for tissue lipid homeostasis, especially in the lung. More recent studies have demonstrated that loss of ABCG1 has wide-ranging consequences and impacts lymphocyte and stem cell proliferation, endothelial cell function, macrophage foam cell formation, as well as insulin secretion from pancreatic β cells. Recent studies have also demonstrated that ABCG1 functions as an intracellular lipid transporter, localizes to intracellular vesicles/endosomes, and that the transmembrane domains are sufficient for localization and transport function. SUMMARY ABCG1 plays a crucial role in maintaining intracellular sterol and lipid homeostasis. Loss of this transporter has significant, cell-type-specific consequences ranging from effects on cellular proliferation, to surfactant production and/or insulin secretion. Elucidation of the mechanisms by which ABCG1 affects intracellular sterol flux/movement should provide important information that may link ABCG1 to diseases of dysregulated tissue lipid homeostasis.
Collapse
Affiliation(s)
- Elizabeth J Tarling
- Departments of Biological Chemistry and Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1737, USA.
| |
Collapse
|
49
|
Abstract
Lysophosphatidic acid (LPA) is a potent bioactive phospholipid. As many other biological active lipids, LPA is an autacoid: it is formed locally on demand, and it acts locally near its site of synthesis. LPA has a plethora of biological activities on blood cells (platelets, monocytes) and cells of the vessel wall (endothelial cells, smooth muscle cells, macrophages) that are all key players in atherosclerotic and atherothrombotic processes. The specific cellular actions of LPA are determined by its multifaceted molecular structures, the expression of multiple G-protein coupled LPA receptors at the cell surface and their diverse coupling to intracellular signalling pathways. Numerous studies have now shown that LPA has thrombogenic and atherogenic actions. Here, we aim to provide a comprehensive, yet concise, thoughtful and critical review of this exciting research area and to pinpoint potential pharmacological targets for inhibiting thrombogenic and atherogenic activities of LPA. We hope that the review will serve to accelerate knowledge of basic and clinical science, and to foster drug development in the field of LPA and atherosclerotic/atherothrombotic diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
50
|
Gursky O. Crystal structure of Δ(185-243)ApoA-I suggests a mechanistic framework for the protein adaptation to the changing lipid load in good cholesterol: from flatland to sphereland via double belt, belt buckle, double hairpin and trefoil/tetrafoil. J Mol Biol 2013; 425:1-16. [PMID: 23041415 PMCID: PMC3534807 DOI: 10.1016/j.jmb.2012.09.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/23/2012] [Accepted: 09/29/2012] [Indexed: 12/18/2022]
Abstract
Apolipoprotein A-I (apoA-I) is the major protein of plasma high-density lipoproteins (HDLs), macromolecular assemblies of proteins and lipids that remove cell cholesterol and protect against atherosclerosis. HDL heterogeneity, large size (7.7-12 nm), and ability to exchange proteins have prevented high-resolution structural analysis. Low-resolution studies showed that two apoA-I molecules form an antiparallel α-helical "double belt" around an HDL particle. The atomic-resolution structure of the C-terminal truncated lipid-free Δ(185-243)apoA-I, determined recently by Mei and Atkinson, provides unprecedented new insights into HDL structure-function. It allows us to propose a molecular mechanism for the adaptation of the full-length protein to increasing lipid load during cholesterol transport. ApoA-I conformations on small, midsize, and large HDLs are proposed based on the tandem α-helical repeats and the crystal structure of Δ(185-243)apoA-I and are validated by comparison with extensive biophysical data reported by many groups. In our models, the central half of the double belt ("constant" segment 66-184) is structurally conserved while the N- and C-terminal half ("variable" segments 1-65 and 185-243) rearranges upon HDL growth. This includes incremental unhinging of the N-terminal bundle around two flexible regions containing G39 and G65 to elongate the belt, along with concerted swing motion of the double belt around G65-P66 and G185-G186 hinges that are aligned on various-size particles, to confer two-dimensional surface curvature to spherical HDLs. The proposed conformational ensemble integrates and improves several existing HDL models. It helps provide a structural framework necessary to understand functional interactions with over 60 other HDL-associated proteins and, ultimately, improve the cardioprotective function of HDL.
Collapse
Affiliation(s)
- Olga Gursky
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|