1
|
Benitez Amaro A, Solanelles Curco A, Garcia E, Julve J, Rives J, Benitez S, Llorente Cortes V. Apolipoprotein and LRP1-Based Peptides as New Therapeutic Tools in Atherosclerosis. J Clin Med 2021; 10:jcm10163571. [PMID: 34441867 PMCID: PMC8396846 DOI: 10.3390/jcm10163571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/17/2022] Open
Abstract
Apolipoprotein (Apo)-based mimetic peptides have been shown to reduce atherosclerosis. Most of the ApoC-II and ApoE mimetics exert anti-atherosclerotic effects by improving lipid profile. ApoC-II mimetics reverse hypertriglyceridemia and ApoE-based peptides such as Ac-hE18A-NH2 reduce cholesterol and triglyceride (TG) levels in humans. Conversely, other classes of ApoE and ApoA-I mimetic peptides and, more recently, ApoJ and LRP1-based peptides, exhibit several anti-atherosclerotic actions in experimental models without influencing lipoprotein profile. These other mimetic peptides display at least one atheroprotective mechanism such as providing LDL stability against mechanical modification or conferring protection against the action of lipolytic enzymes inducing LDL aggregation in the arterial intima. Other anti-atherosclerotic effects exerted by these peptides also include protection against foam cell formation and inflammation, and induction of reverse cholesterol transport. Although the underlying mechanisms of action are still poorly described, the recent findings suggest that these mimetics could confer atheroprotection by favorably influencing lipoprotein function rather than lipoprotein levels. Despite the promising results obtained with peptide mimetics, the assessment of their stability, atheroprotective efficacy and tissue targeted delivery are issues currently under progress.
Collapse
Affiliation(s)
- Aleyda Benitez Amaro
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.A.); (E.G.)
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain;
| | | | - Eduardo Garcia
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.A.); (E.G.)
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain;
| | - Josep Julve
- Metabolic Basis of Cardiovascular Risk Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Jose Rives
- Biochemistry Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, 08016 Barcelona, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Correspondence: (S.B.); or (V.L.C.)
| | - Vicenta Llorente Cortes
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.A.); (E.G.)
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain;
- CIBERCV, Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (S.B.); or (V.L.C.)
| |
Collapse
|
2
|
Luo H, Lu L, Liu N, Li Q, Yang X, Zhang Z. Curcumin loaded sub-30 nm targeting therapeutic lipid nanoparticles for synergistically blocking nasopharyngeal cancer growth and metastasis. J Nanobiotechnology 2021; 19:224. [PMID: 34320999 PMCID: PMC8317404 DOI: 10.1186/s12951-021-00966-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/19/2021] [Indexed: 01/07/2023] Open
Abstract
Systemic chemotherapy is still the primary treatment for advanced-stage nasopharyngeal carcinoma (NPC), but only limited therapeutic success has been achieved in the past decade because of drug resistance and systemic toxicity. Curcumin (Cur) is an effective alternative to chemotherapeutics because it showed remarkable therapeutic potential in the treatment of NPC. However, lack of tissue specificity and poor penetration in solid tumors are the major obstacles to effective therapy. Therefore, in this work, a self-assembled sub-30 nm therapeutic lipid nanoparticle loaded with Cur, named as Cur@α-NTP-LN, was constructed, specifically targeting scavenger receptor class B member 1 (SR-B1) and enhancing its therapeutic effects on NPC in vivo. Our results showed that Cur@α-NTP-LNs were effective and superior to free Cur on NPC cell-specific targeting, suppressing cell proliferation and inducing cell apoptosis. In vivo and ex vivo optical imaging revealed that Cur@α-NTP-LNs exerted high targeting efficiency, specifically accumulating in NPC xenograft tumors and delivering Cur into the tumor center after systemic administration. Furthermore, Cur@α-NTP-LNs exhibited a remarkable inhibitory effect on the growth of NPC subcutaneous tumors, with over 71 and 47% inhibition compared to Cur- and α-NTP-LNs-treated groups, respectively. In addition, Cur@α-NTP-LNs almost blocked NPC metastasis in a lung metastasis model of NPC and significantly improved the survival rate. Thus, the sub-30 nm Cur@α-NTP-LNs enhanced the solubility of Cur and demonstrated the ability of targeted Cur delivery into the center of the solid NPC tumor, performing synergistic inhibitory effects on the growth of NPC tumor and its metastasis with high efficiency. ![]()
Collapse
Affiliation(s)
- Haiming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China. .,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ni Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qingqing Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China. .,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
3
|
Role of Short Chain Fatty Acids and Apolipoproteins in the Regulation of Eosinophilia-Associated Diseases. Int J Mol Sci 2021; 22:ijms22094377. [PMID: 33922158 PMCID: PMC8122716 DOI: 10.3390/ijms22094377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Eosinophils are key components of our host defense and potent effectors in allergic and inflammatory diseases. Once recruited to the inflammatory site, eosinophils release their cytotoxic granule proteins as well as cytokines and lipid mediators, contributing to parasite clearance but also to exacerbation of inflammation and tissue damage. However, eosinophils have recently been shown to play an important homeostatic role in different tissues under steady state. Despite the tremendous progress in the treatment of eosinophilic disorders with the implementation of biologics, there is an unmet need for novel therapies that specifically target the cytotoxic effector functions of eosinophils without completely depleting this multifunctional immune cell type. Recent studies have uncovered several endogenous molecules that decrease eosinophil migration and activation. These include short chain fatty acids (SCFAs) such as butyrate, which are produced in large quantities in the gastrointestinal tract by commensal bacteria and enter the systemic circulation. In addition, high-density lipoprotein-associated anti-inflammatory apolipoproteins have recently been shown to attenuate eosinophil migration and activation. Here, we focus on the anti-pathogenic properties of SCFAs and apolipoproteins on eosinophil effector function and provide insights into the potential use of SCFAs and apolipoproteins (and their mimetics) as effective agents to combat eosinophilic inflammation.
Collapse
|
4
|
Pillai JA, Bena J, Bekris LM, Foldvary-Schaefer N, Heinzinger C, Rao S, Rao SM, Leverenz JB, Mehra R. Unique Sleep and Circadian Rhythm Dysfunction Neuroinflammatory and Immune Profiles in Alzheimer's Disease with Mild Cognitive Impairment. J Alzheimers Dis 2021; 81:487-492. [PMID: 33814445 PMCID: PMC8179975 DOI: 10.3233/jad-201573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sleep dysfunction has been identified in the pathophysiology of Alzheimer's disease (AD); however, the role and mechanism of circadian rhythm dysfunction is less well understood. In a well-characterized cohort of patients with AD at the mild cognitive impairment stage (MCI-AD), we identify that circadian rhythm irregularities were accompanied by altered humoral immune responses detected in both the cerebrospinal fluid and plasma as well as alterations of cerebrospinal fluid biomarkers of neurodegeneration. On the other hand, sleep disruption was more so associated with abnormalities in circulating markers of immunity and inflammation and decrements in cognition.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Catherine Heinzinger
- Sleep Disorders Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sujata Rao
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Stephen M. Rao
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - Reena Mehra
- Sleep Disorders Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
5
|
Peck MJ, Sanders EB, Scherer G, Lüdicke F, Weitkunat R. Review of biomarkers to assess the effects of switching from cigarettes to modified risk tobacco products. Biomarkers 2018; 23:213-244. [PMID: 29297706 DOI: 10.1080/1354750x.2017.1419284] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Context: One approach to reducing the harm caused by cigarette smoking, at both individual and population level, is to develop, assess and commercialize modified risk alternatives that adult smokers can switch to. Studies to demonstrate the exposure and risk reduction potential of such products generally involve the measuring of biomarkers, of both exposure and effect, sampled in various biological matrices.Objective: In this review, we detail the pros and cons for using several biomarkers as indicators of effects of changing from conventional cigarettes to modified risk products.Materials and methods: English language publications between 2008 and 2017 were retrieved from PubMed using the same search criteria for each of the 25 assessed biomarkers. Nine exclusion criteria were applied to exclude non-relevant publications.Results: A total of 8876 articles were retrieved (of which 7476 were excluded according to the exclusion criteria). The literature indicates that not all assessed biomarkers return to baseline levels following smoking cessation during the study periods but that nine had potential for use in medium to long-term studies.Discussion and conclusion: In clinical studies, it is important to choose biomarkers that show the biological effect of cessation within the duration of the study.
Collapse
Affiliation(s)
| | | | | | - Frank Lüdicke
- Research & Development, Philip Morris International, Neuchâtel, Switzerland
| | - Rolf Weitkunat
- Research & Development, Philip Morris International, Neuchâtel, Switzerland
| |
Collapse
|
6
|
Ding Y, Li B, Tian F, Zhou S, Chen Y. Effects of blood lipid stability on progression of carotid atherosclerosis. Pak J Med Sci 2017; 33:599-602. [PMID: 28811778 PMCID: PMC5510110 DOI: 10.12669/pjms.333.12593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objective: To evaluate the effects of blood lipid stability on progression of carotid atherosclerosis. Methods: A total of 416 patients who had physical examination in our hospital annually from January 2010 to December 2015 were selected and divided into a progression group (n=216) and a non-progression group (n=200) according to the intima-media thickness measured by carotid ultrasound. The levels of lipid-related parameters within five years were retrospectively analyzed to calculate the smoothness index (SI = x±/s). Results: The cross-sectional TG, HDL-C, ApoAI, ApoB, ApoE and Lpa levels were similar in the two groups (p>0.05). The non-progression group had significantly higher TC ((4.15±0.82 vs. 4.50±1.04) mmol/L) and LDL-C ((2.53±0.76 vs. 2.99±1.03) mmol/L) levels than those of the progression group (p<0.05). The progression group had significantly lower TC SI (5.29±1.28 vs. 5.65±1.76), TG SI (2.13±0.71 vs. 2.79±0.82), LDL-C SI (3.66±1.17 vs. 4.36±1.58), ApoB SI (3.37±0.88 vs. 3.62±0.95) and Lpa SI (1.53±0.49 vs. 1.62±0.43) than those of the non-progression group (p<0.05). Conclusion: Compared with cross-sectional results, SI was better correlated with the progression of atherosclerosis. The progression group had lower SI values.
Collapse
Affiliation(s)
- Yu Ding
- Yu Ding, Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Li
- Bo Li, Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Feng Tian
- Feng Tian, Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Shanshan Zhou
- Shanshan Zhou, Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yundai Chen
- Yundai Chen, Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
7
|
Zhao Y, Leman LJ, Search DJ, Garcia RA, Gordon DA, Maryanoff BE, Ghadiri MR. Self-Assembling Cyclic d,l-α-Peptides as Modulators of Plasma HDL Function. A Supramolecular Approach toward Antiatherosclerotic Agents. ACS CENTRAL SCIENCE 2017; 3:639-646. [PMID: 28691076 PMCID: PMC5492419 DOI: 10.1021/acscentsci.7b00154] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Indexed: 05/26/2023]
Abstract
There is great interest in developing new modes of therapy for atherosclerosis to treat coronary heart disease and stroke, particularly ones that involve modulation of high-density lipoproteins (HDLs). Here, we describe a new supramolecular chemotype for altering HDL morphology and function. Guided by rational design and SAR-driven peptide sequence enumerations, we have synthesized and determined the HDL remodeling activities of over 80 cyclic d,l-α-peptides. We have identified a few distinct sequence motifs that are effective in vitro in remodeling human and mouse plasma HDLs to increase the concentration of lipid-poor pre-beta HDLs, which are key initial acceptors of cholesterol in the reverse cholesterol transport (RCT) process, and concomitantly promote cholesterol efflux from macrophage cells. Functional assays with various control peptides, such as scrambled sequences, linear and enantiomeric cyclic peptide variants, and backbone-modified structures that limit peptide self-assembly, provide strong support for the supramolecular mode of action. Importantly, when the lead cyclic peptide c[wLwReQeR] was administered to mice (ip), it also promoted the formation of small, lipid-poor HDLs in vivo, displayed good plasma half-life (∼6 h), did not appear to have adverse side effects, and exerted potent anti-inflammatory effects in an acute in vivo inflammation assay. Given that previously reported HDL remodeling peptides have been based on α-helical apoA-I mimetic architectures, the present study, involving a new structural class, represents a promising step toward new potential therapeutics to combat atherosclerosis.
Collapse
Affiliation(s)
- Yannan Zhao
- 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
| | - 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
| | - Debra J. Search
- Cardiovascular
Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, United States
| | - Ricardo A. Garcia
- Cardiovascular
Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, United States
| | - David A. Gordon
- Cardiovascular
Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, United States
| | - Bruce E. Maryanoff
- 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
| | - M. Reza Ghadiri
- 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
|
8
|
Correlations Between Serum Apolipoprotein A-I and Formation of Vocal Cord Polyp. J Voice 2017; 31:380.e1-380.e6. [DOI: 10.1016/j.jvoice.2016.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 11/17/2022]
|
9
|
Jin X, Sviridov D, Liu Y, Vaisman B, Addadi L, Remaley AT, Kruth HS. ABCA1 (ATP-Binding Cassette Transporter A1) Mediates ApoA-I (Apolipoprotein A-I) and ApoA-I Mimetic Peptide Mobilization of Extracellular Cholesterol Microdomains Deposited by Macrophages. Arterioscler Thromb Vasc Biol 2016; 36:2283-2291. [PMID: 27758769 DOI: 10.1161/atvbaha.116.308334] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/02/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We examined the function of ABCA1 (ATP-binding cassette transporter A1) in ApoA-I (apolipoprotein A-I) mobilization of cholesterol microdomains deposited into the extracellular matrix by cholesterol-enriched macrophages. We have also determined whether an ApoA-I mimetic peptide without and with complexing to sphingomyelin can mobilize macrophage-deposited cholesterol microdomains. APPROACH AND RESULTS Extracellular cholesterol microdomains deposited by cholesterol-enriched macrophages were detected with a monoclonal antibody, 58B1. ApoA-I and an ApoA-I mimetic peptide 5A mobilized cholesterol microdomains deposited by ABCA1+/+ macrophages but not by ABCA1-/- macrophages. In contrast, ApoA-I mimetic peptide 5A complexed with sphingomyelin could mobilize cholesterol microdomains deposited by ABCA1-/- macrophages. CONCLUSIONS Our findings show that a unique pool of extracellular cholesterol microdomains deposited by macrophages can be mobilized by both ApoA-I and an ApoA-I mimetic peptide but that mobilization depends on macrophage ABCA1. It is known that ABCA1 complexes ApoA-I and ApoA-I mimetic peptide with phospholipid, a cholesterol-solubilizing agent, explaining the requirement for ABCA1 in extracellular cholesterol microdomain mobilization. Importantly, ApoA-I mimetic peptide already complexed with phospholipid can mobilize macrophage-deposited extracellular cholesterol microdomains even in the absence of ABCA1.
Collapse
Affiliation(s)
- Xueting Jin
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.)
| | - Denis Sviridov
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.)
| | - Ying Liu
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.)
| | - Boris Vaisman
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.)
| | - Lia Addadi
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.)
| | - Alan T Remaley
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.)
| | - Howard S Kruth
- From the Experimental Atherosclerosis Section (X.J., Y.L., H.S.K.) and Lipoprotein Metabolism Section (D.S., B.V., A.T.R.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel (L.A.).
| |
Collapse
|
10
|
High-density Lipoprotein and Inflammation and Its Significance to Atherosclerosis. Am J Med Sci 2016; 352:408-415. [DOI: 10.1016/j.amjms.2016.06.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 06/06/2016] [Accepted: 06/24/2016] [Indexed: 01/09/2023]
|
11
|
Babintseva YD, Camont L, Chapman J, Lhomme M, Karagodin VP, Kontush A, Orekhov AN. The biological activity of high-density lipoprotein fractions and their role in the development of cardiovascular diseases. TERAPEVT ARKH 2016. [DOI: 10.17116/terarkh2016889111-118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Increasing the human plasma concentration of high-density lipoproteins (HDL) may be part of strategy for control of cardiovascular diseases (CVD). HDL particles vary in their structure, metabolism, and biological activity. The review describes major HDL fractions (subpopulations) and discusses new findings on the antiatherogenic properties of HDL particles. The whole spectrum of HDL fractions, small, dense, protein-rich lipoproteins, has atheroprotective properties that are determined by the presence of specialized groups of proteins and lipids; however, this activity may be decreased in atherogenic lesion. Comprehensive structural and compositional analysis of HDL may provide key information to identify the fractions that have characteristic biological properties and lose their functionality in CVD. These fractions may be also biomarkers for the risk of CVD and hence represent pharmacological targets.
Collapse
|
12
|
PAPPIANI C, DAMASCENO NRT. Impacto da suplementação com ácidos graxos ômega-3 nas subfrações da lipoproteína de alta densidade de indivíduos tabagistas. REV NUTR 2016. [DOI: 10.1590/1678-98652016000400006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
RESUMO Objetivo Avaliar o efeito da suplementação com ômega-3 nas subfrações das lipoproteínas de alta densidade em indivíduos tabagistas. Métodos Ensaio clínico, randomizado, duplo-cego. Foi selecionada uma amostra com 33 tabagistas, de ambos os sexos, com idade entre 30 e 60 anos, suplementados com ômega-3 (n=17) ou placebo (ácidos graxos ômega-6, n=16) por dois meses. As subfrações das lipoproteínas de alta densidade foram analisadas pelo sistema Lipoprint. Os testes estatísticos foram realizados com o auxílio do programa Statistical Package for the Social Sciences, versão 20.0. Resultados A média de idade foi 49 anos, com predominância da raça branca. Após a intervenção, o grupo ômega-3 modificou positivamente o perfil lipídico e as subfrações das lipoproteínas de alta densidade dos tabagistas. Nos modelos de regressão linear testados, o percentual de ácido docosahexaenoico plasmático apresentou associações negativas com o percentual das lipoproteínas de alta densidade-pequena. Conclusão A suplementação com ômega-3 está associada a uma alteração favorável na distribuição das subfrações das lipoproteínas de alta densidade, aumentando as lipoproteínas de alta densidade-grande e diminuindo as lipoproteínas de alta densidade-pequena. Isso reforça a importância do ômega-3 na saúde cardiovascular de indivíduos tabagistas.
Collapse
|
13
|
Sandim V, Pereira DDA, Kalume DE, Oliveira-Carvalho AL, Ornellas AA, Soares MR, Alves G, Zingali RB. Proteomic analysis reveals differentially secreted proteins in the urine from patients with clear cell renal cell carcinoma. Urol Oncol 2015; 34:5.e11-25. [PMID: 26420021 DOI: 10.1016/j.urolonc.2015.07.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/04/2015] [Accepted: 07/26/2015] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the differentially secreted protein profile in the urine from patients with clear cell renal cell carcinoma (ccRCC) using mass spectrometry-based methods. Urine composition can reflect kidney physiology and can be used to detect markers for renal diseases. Moreover, characterization of the secretome is likely to assist in the investigation of new drugs for biological targets and diagnose the ccRCC at an early stage. METHODS AND MATERIALS Urine samples from patients were divided according to Fuhrman degree (FI-IV), which was associated with the cellular differentiation as good prognosis (GP) and poor prognosis (PP). Healthy individuals were used as the control group (CG). We used both qualitative and quantitative mass spectrometry-based analyses that involved the following approaches: 1-dimensional gel electrophoresis combined with liquid chromatography mass spectrometry in tandem (1DE LC-MS/MS), in-solution digestion combined with label-free 1-dimensional LC-MS(E) (1D LC-MS(E)), and bidimensional gel electrophoresis combined with matrix-assisted laser desorption/ionization time of flight in tandem (2DE MALDI-TOF/TOF) or combined with LC-MS/MS. RESULTS All the strategies allowed the identification of 354 proteins from the CG, GP, and PP groups. Qualitative experiments using 1DE LC-MS/MS analysis detected different protein profiles, and 224 proteins were identified in all groups. The label-free MS(E) quantitative analysis identified 113 proteins and generated novel information on secreted protein profiles, including 49 up-secreted proteins in the urine from patients with ccRCC and 40 down-secreted proteins related to the CG. Proteins such as kininogen-1, uromodulin, apolipoprotein D, polyubiquitin, and CD59 glycoprotein were down secreted according to the groups CG>GP>PP. In contrast, apolipoprotein A, fibrinogen, and haptoglobin were up secreted in patient groups. The same expression profile observed for kininogen-1, apolipoprotein D, fibrinogen, and haptoglobin was corroborated by 2DE LC-MS/MS or 2DE MALDI-TOF/TOF analyses. These 2 strategies also showed 13 differentially secreted proteins among the 3 groups. CONCLUSIONS The proteins kininogen-1, apolipoprotein D, fibrinogen, and haptoglobin presented similar quantitative protein profiles according to MS(E) and 2DE approaches. The latter proteins were up secreted and the former ones were down-regulated. The strategies used proved to be valuable in identifying proteins that were differentially secreted in urine from patients with RCC.
Collapse
Affiliation(s)
- Vanessa Sandim
- Unidade de Espectrometria de Massas e Proteômica (UEMP), Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Instituto Nacional de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Laboratório de Genética Aplicada, Serviço de Hematologia, Instituto Nacional de Câncer-INCA, Rio de Janeiro, Brazil; Laboratório de Marcadores Circulantes, Departamento de Patologia, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro - UERJ, Rio de Janeiro, Brazil
| | - Denise de Abreu Pereira
- Unidade de Espectrometria de Massas e Proteômica (UEMP), Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Instituto Nacional de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Laboratório de Genética Aplicada, Serviço de Hematologia, Instituto Nacional de Câncer-INCA, Rio de Janeiro, Brazil; Coordenação Geral de Ensino e Pesquisa, Instituto Nacional de Câncer-INCA, Rio de Janeiro, Brazil
| | - Dário Eluan Kalume
- Unidade de Espectrometria de Massas e Proteômica (UEMP), Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Laboratório Interdisciplinar de Pesquisas Médicas (LIPMED), Instituto Oswaldo Cruz - IOC, Fundação Oswaldo Cruz - FIOCRUZ, Rio de Janeiro, Brazil
| | - Ana Lucia Oliveira-Carvalho
- Unidade de Espectrometria de Massas e Proteômica (UEMP), Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Instituto Nacional de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil
| | - Antonio Augusto Ornellas
- Serviço de Urologia, Instituto Nacional de Câncer - INCA, Rio de Janeiro, Brazil; Serviço de Urologia, Hospital Mário Kroeff, Rio de Janeiro, Brazil
| | - Marcia Regina Soares
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil
| | - Gilda Alves
- Laboratório de Genética Aplicada, Serviço de Hematologia, Instituto Nacional de Câncer-INCA, Rio de Janeiro, Brazil; Laboratório de Marcadores Circulantes, Departamento de Patologia, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro - UERJ, Rio de Janeiro, Brazil; Coordenação Geral de Ensino e Pesquisa, Instituto Nacional de Câncer-INCA, Rio de Janeiro, Brazil.
| | - Russolina Benedeta Zingali
- Unidade de Espectrometria de Massas e Proteômica (UEMP), Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil; Instituto Nacional de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Amar MJA, Sakurai T, Sakurai-Ikuta A, Sviridov D, Freeman L, Ahsan L, Remaley AT. A novel apolipoprotein C-II mimetic peptide that activates lipoprotein lipase and decreases serum triglycerides in apolipoprotein E-knockout mice. J Pharmacol Exp Ther 2015; 352:227-35. [PMID: 25395590 PMCID: PMC4293430 DOI: 10.1124/jpet.114.220418] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 11/11/2014] [Indexed: 12/16/2022] Open
Abstract
Apolipoprotein A-I (apoA-I) mimetic peptides are currently being developed as possible new agents for the treatment of cardiovascular disease based on their ability to promote cholesterol efflux and their other beneficial antiatherogenic properties. Many of these peptides, however, have been reported to cause transient hypertriglyceridemia due to inhibition of lipolysis by lipoprotein lipase (LPL). We describe a novel bihelical amphipathic peptide (C-II-a) that contains an amphipathic helix (18A) for binding to lipoproteins and stimulating cholesterol efflux as well as a motif based on the last helix of apolipoprotein C-II (apoC-II) that activates lipolysis by LPL. The C-II-a peptide promoted cholesterol efflux from ATP-binding cassette transporter ABCA1-transfected BHK cells similar to apoA-I mimetic peptides. Furthermore, it was shown in vitro to be comparable to the full-length apoC-II protein in activating lipolysis by LPL. When added to serum from a patient with apoC-II deficiency, it restored normal levels of LPL-induced lipolysis and also enhanced lipolysis in serum from patients with type IV and V hypertriglyceridemia. Intravenous injection of C-II-a (30 mg/kg) in apolipoprotein E-knockout mice resulted in a significant reduction of plasma cholesterol and triglycerides of 38 ± 6% and 85 ± 7%, respectively, at 4 hours. When coinjected with the 5A peptide (60 mg/kg), the C-II-a (30 mg/kg) peptide was found to completely block the hypertriglyceridemic effect of the 5A peptide in C57Bl/6 mice. In summary, C-II-a is a novel peptide based on apoC-II, which promotes cholesterol efflux and lipolysis and may therefore be useful for the treatment of apoC-II deficiency and other forms of hypertriglyceridemia.
Collapse
Affiliation(s)
- Marcelo J A Amar
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Toshihiro Sakurai
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Akiko Sakurai-Ikuta
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Denis Sviridov
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lita Freeman
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lusana Ahsan
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
15
|
Montecucco F, Favari E, Norata GD, Ronda N, Nofer JR, Vuilleumier N. Impact of systemic inflammation and autoimmune diseases on apoA-I and HDL plasma levels and functions. Handb Exp Pharmacol 2015; 224:455-82. [PMID: 25522998 DOI: 10.1007/978-3-319-09665-0_14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cholesterol of high-density lipoproteins (HDLs) and its major proteic component, apoA-I, have been widely investigated as potential predictors of acute cardiovascular (CV) events. In particular, HDL cholesterol levels were shown to be inversely and independently associated with the risk of acute CV diseases in different patient populations, including autoimmune and chronic inflammatory disorders. Some relevant and direct anti-inflammatory activities of HDL have been also recently identified targeting both immune and vascular cell subsets. These studies recently highlighted the improvement of HDL function (instead of circulating levels) as a promising treatment strategy to reduce inflammation and associated CV risk in several diseases, such as systemic lupus erythematosus and rheumatoid arthritis. In these diseases, anti-inflammatory treatments targeting HDL function might improve both disease activity and CV risk. In this narrative review, we will focus on the pathophysiological relevance of HDL and apoA-I levels/functions in different acute and chronic inflammatory pathophysiological conditions.
Collapse
Affiliation(s)
- Fabrizio Montecucco
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1211, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
16
|
Gerster R, Eloranta JJ, Hausmann M, Ruiz PA, Cosin-Roger J, Terhalle A, Ziegler U, Kullak-Ublick GA, von Eckardstein A, Rogler G. Anti-inflammatory Function of High-Density Lipoproteins via Autophagy of IκB Kinase. Cell Mol Gastroenterol Hepatol 2014; 1:171-187.e1. [PMID: 28247863 PMCID: PMC5301135 DOI: 10.1016/j.jcmgh.2014.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Plasma levels of high-density lipoprotein (HDL) cholesterol are frequently found decreased in patients with inflammatory bowel disease (IBD). Therefore, and because HDL exerts anti-inflammatory activities, we investigated whether HDL and its major protein component apolipoprotein A-I (apoA-I) modulate mucosal inflammatory responses in vitro and in vivo. METHODS The human intestinal epithelial cell line T84 was used as the in vitro model for measuring the effects of HDL on the expression and secretion of tumor necrosis factor (TNF), interleukin-8 (IL-8), and intracellular adhesion molecule (ICAM). Nuclear factor-κB (NF-κB)-responsive promoter activity was studied by dual luciferase reporter assays. Mucosal damage from colitis induced by dextran sodium sulphate (DSS) and 2,4,6-trinitrobenzenesulfonic acid (TNBS) was scored by colonoscopy and histology in apoA-I transgenic (Tg) and apoA-I knockout (KO) and wild-type (WT) mice. Myeloperoxidase (MPO) activity and TNF and ICAM expression were determined in intestinal tissue samples. Autophagy was studied by Western blot analysis, immunofluorescence, and electron microscopy. RESULTS HDL and apoA-I down-regulated TNF-induced mRNA expression of TNF, IL-8, and ICAM, as well as TNF-induced NF-κB-responsive promoter activity. DSS/TNBS-treated apoA-I KO mice displayed increased mucosal damage upon both colonoscopy and histology, increased intestinal MPO activity and mRNA expression of TNF and ICAM as compared with WT and apoA-I Tg mice. In contrast, apoA-I Tg mice showed less severe symptoms monitored by colonoscopy and MPO activity in both the DSS and TNBS colitis models. In addition, HDL induced autophagy, leading to recruitment of phosphorylated IκB kinase to the autophagosome compartment, thereby preventing NF-κB activation and induction of cytokine expression. CONCLUSIONS Taken together, the in vitro and in vivo findings suggest that HDL and apoA-I suppress intestinal inflammation via autophagy and are potential therapeutic targets for the treatment of IBD.
Collapse
Key Words
- 3-MA, 3-methyl adenine
- ApoA-I, apolipoprotein A-I
- Apolipoprotein A-I
- Autophagy
- CD, Crohn’s disease
- DAPI, 4′,6-diamidino-2-phenylindole
- DSS, dextran sodium sulphate
- EMSA, electrophoretic mobility shift assay
- HDL, high-density lipoprotein
- IBD, inflammatory bowel disease
- ICAM, intracellular adhesion molecule
- IL, interleukin
- Inflammatory Bowel Disease
- KO, knockout
- LC3II, light chain 3 II
- MEICS, murine endoscopic index of colitis severity
- MPO, myeloperoxidase
- NF-κB
- NF-κB, nuclear factor κB
- PBS, phosphate-buffered saline
- PFA, paraformaldehyde
- PI-3, phosphatidylinositol-3
- RT-PCR, real-time polymerase chain reaction
- TNBS, 2,4,6-trinitrobenzenesulfonic acid
- TNF, tumor necrosis factor
- Tg, transgenic
- WT, wild type
- mTOR, the mammalian target of rapamycin
- p-IKK, phosphorylated IκB kinase
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Ragam Gerster
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Schlieren, Switzerland
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Jyrki J. Eloranta
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Schlieren, Switzerland
| | - Martin Hausmann
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Pedro A. Ruiz
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jesus Cosin-Roger
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Anne Terhalle
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Centre for Microscopy and Image Analysis, University Hospital Zurich, Zurich, Switzerland
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Schlieren, Switzerland
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Correspondence Address correspondence to: Gerhard Rogler, MD, PhD, Division of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland. fax: +41-0-44-255-9497.
| |
Collapse
|
17
|
Wang F, Ye P. Improving heart function by modulating myocardiocyte autophagy: a possible novel mechanism for cardiovascular protection of high-density lipoprotein. Lipids Health Dis 2014; 13:163. [PMID: 25339382 PMCID: PMC4216853 DOI: 10.1186/1476-511x-13-163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High-density lipoprotein (HDL) has been shown to confer cardiovascular protection in clinical and epidemiologic studies. Emerging evidence suggests that many of the cardioprotective functions of HDL may be due to the phospholipid sphingosine-1-phosphate (S1P). PRESENTATION OF THE HYPOTHESIS HDL-S1P binds to S1P receptors in the heart, activating PI3K/Akt signaling and myocyte survival. PI3K/Akt is a classic signaling modulator of autophagy. Excessive autophagy due to cell death and cardiomyocyte loss may contribute to impaired heart function during pressure overload-induced heart failure. Therefore, we hypothesize that HDL-S1P may suppress excessive autophagy of cardiomyocytes through activation of PI3K/Akt signaling. Further, reconstituted HDL (including S1P) may protect heart function during pressure overload-induced heart failure. TESTING THE HYPOTHESIS We will design the following experiments to test this hypothesis. (1) We will treat cells and mice with PI-3 kinase inhibitors to examine if HDL-S1P downregulates expression of Autophagy-related genes (ATGs) and proteins via activation of PI3K/Akt signaling. (2) We will use siRNA against S1P receptors or inhibitors of S1P receptors to determine which types of S1P receptors participate in this mechanism. (3) We will also examine if reconstituted HDL (including S1P) improves heart function during pressure overload-induced heart failure by suppressing excessive autophagy of cardiomyocytes through activation of PI3K/Akt signaling. IMPLICATIONS OF THE HYPOTHESIS Understanding the autophagy signaling pathway modulated by HDL-S1P will make a major contribution to the field by identifying a novel mechanism for cardiovascular protection of high-density lipoprotein. Further, using reconstituted HDL to improve heart function would provide a novel therapeutic approach for pressure overload-induced heart failure.
Collapse
Affiliation(s)
| | - Ping Ye
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
18
|
Zhao Y, Black AS, Bonnet DJ, Maryanoff BE, Curtiss LK, Leman LJ, Ghadiri MR. In vivo efficacy of HDL-like nanolipid particles containing multivalent peptide mimetics of apolipoprotein A-I. J Lipid Res 2014; 55:2053-63. [PMID: 24975585 DOI: 10.1194/jlr.m049262] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We have observed that molecular constructs based on multiple apoA-I mimetic peptides attached to a branched scaffold display promising anti-atherosclerosis functions in vitro. Building on these promising results, we now describe chronic in vivo studies to assess anti-atherosclerotic efficacy of HDL-like nanoparticles assembled from a trimeric construct, administered over 10 weeks either ip or orally to LDL receptor-null mice. When dosed ip, the trimer-based nanolipids markedly reduced plasma LDL-cholesterol levels by 40%, unlike many other apoA-I mimetic peptides, and were substantially atheroprotective. Surprisingly, these nanoparticles were also effective when administered orally at a dose of 75 mg/kg, despite the peptide construct being composed of l-amino acids and being undetectable in the plasma. The orally administered nanoparticles reduced whole aorta lesion areas by 55% and aortic sinus lesion volumes by 71%. Reductions in plasma cholesterol were due to the loss of non-HDL lipoproteins, while plasma HDL-cholesterol levels were increased. At a 10-fold lower oral dose, the nanoparticles were marginally effective in reducing atherosclerotic lesions. Intriguingly, analogous results were obtained with nanolipids of the corresponding monomeric peptide. These nanolipid formulations provide an avenue for developing orally efficacious therapeutic agents to manage atherosclerosis.
Collapse
Affiliation(s)
- Yannan Zhao
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| | - Audrey S Black
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| | - David J Bonnet
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| | - Bruce E Maryanoff
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| | - Linda K Curtiss
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| | - Luke J Leman
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| | - M Reza Ghadiri
- Departments of Chemistry and Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
19
|
Xu XR, Zhong L, Huang BL, Wei YH, Zhou X, Wang L, Wang FQ. Comparative proteomic analysis of plasma proteins in patients with age-related macular degeneration. Int J Ophthalmol 2014; 7:256-63. [PMID: 24790867 DOI: 10.3980/j.issn.2222-3959.2014.02.12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 12/18/2013] [Indexed: 09/26/2022] Open
Abstract
AIM To find the significant altered proteins in age-related macular degeneration (AMD) patients as potential biomarkers of AMD. METHODS A comparative analysis of the protein pattern of AMD patients versus healthy controls was performed by means of proteomic analysis using two-dimensional gel electrophoresis followed by protein identification with MALDI TOF/TOF mass spectrometry. RESULTS We identified 28 proteins that were significantly altered with clinical relevance in AMD patients. These proteins were involved in a wide range of biological functions including immune responses, growth cytokines, cell fate determination, wound healing, metabolism, and anti-oxidance. CONCLUSION These results demonstrate the capacity of proteomic analysis of AMD patient plasma. In addition to the utility of this approach for biomarker discovery, identification of alterations in endogenous proteins in the plasma of AMD patient could improve our understanding of the disease pathogenesis.
Collapse
Affiliation(s)
- Xin-Rong Xu
- Department of Ophthalmology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Lu Zhong
- Department of Ophthalmology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Bing-Lin Huang
- Department of Ophthalmology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yuan-Hua Wei
- Department of Clinical Laboratory, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Xin Zhou
- Department of Ophthalmology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Ling Wang
- Analytical Instrumentation Center, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Fu-Qiang Wang
- Analytical Instrumentation Center, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
20
|
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
|
21
|
Zhao Y, Imura T, Leman LJ, Curtiss LK, Maryanoff BE, Ghadiri MR. Mimicry of high-density lipoprotein: functional peptide-lipid nanoparticles based on multivalent peptide constructs. J Am Chem Soc 2013; 135:13414-24. [PMID: 23978057 DOI: 10.1021/ja404714a] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We describe an approach for engineering peptide-lipid nanoparticles that function similarly to high-density lipoprotein (HDL). Branched, multivalent constructs, bearing multiple 23- or 16-amino-acid peptides, were designed, synthesized, and combined with phospholipids to produce nanometer-scale discoidal HDL-like particles. A variety of biophysical techniques were employed to characterize the constructs, including size exclusion chromatography, analytical ultracentrifuge sedimentation, circular dichroism, transmission electron microscopy, and fluorescence spectroscopy. The nanoparticles functioned in vitro (human and mouse plasma) and in vivo (mice) to rapidly remodel large native HDLs into small lipid-poor HDL particles, which are key acceptors of cholesterol in reverse cholesterol transport. Fluorescent labeling studies showed that the constituents of the nanoparticles readily distributed into native HDLs, such that the peptide constructs coexisted with apolipoprotein A-I (apoA-I), the main structural protein in HDLs. Importantly, nanolipid particles containing multivalent peptides promoted efficient cellular cholesterol efflux and were functionally superior to those derived from monomeric apoA-I mimetic peptides. The multivalent peptide-lipid nanoparticles were also remarkably stable toward enzymatic digestion in vitro and displayed long half-lives and desirable pharmacokinetic profiles in mice, providing a real practical advantage over previously studied linear or tandem helical peptides. Encouragingly, a two-week exploratory efficacy study in a widely used animal model for atherosclerosis research (LDLr-null mice) using nanoparticles constructed from a trimeric peptide demonstrated an exceptional 50% reduction in the plasma total cholesterol levels compared to the control group. Altogether, the studies reported here point to an attractive avenue for designing synthetic, HDL-like nanoparticles, with potential for treating atherosclerosis.
Collapse
Affiliation(s)
- Yannan Zhao
- Department of Chemistry, ‡Department of Immunology and Microbial Science, and §The Skaggs Institute for Chemical Biology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | | | | | | | |
Collapse
|
22
|
Insulin resistance, serum visfatin, and adiponectin levels are associated with metabolic disorders in chronic hepatitis C virus-infected patients. Eur J Gastroenterol Hepatol 2013; 25:935-41. [PMID: 23470357 DOI: 10.1097/meg.0b013e32835fa988] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The aim of this study was to determine the serum levels of visfatin, adiponectin, and insulin resistance (IR) in patients with hepatitis C virus (HCV) infection and their relations to the biochemical markers of hepatitis C. MATERIALS AND METHODS This study was carried out on 40 HCV-infected patients and 40 sex/age/BMI-matched healthy adults. Lipid profile, liver function tests, IR, serum adiponectin, and visfatin of all patients were examined. Correlations between IR, adiponectin, visfatin, and other variables were analyzed. RESULTS The levels of visfatin and adiponectin were significantly lower in HCV patients compared with healthy controls. However, IR of HCV patients were higher than those of healthy controls. IR was significantly correlated to triglycerides, visfatin was closely related to low-density lipoprotein cholesterol, whereas adiponectin was associated with high-density lipoprotein cholesterol. These results suggest that IR, serum visfatin, and adiponectin levels are associated with metabolic disorders in chronic HCV-infected patients. CONCLUSION IR, adiponectin, and visfatin were related to several metabolic markers of HCV, suggesting the characteristics of HCV-related metabolic abnormalities.
Collapse
|
23
|
Filippatos TD, Elisaf MS. High density lipoprotein and cardiovascular diseases. World J Cardiol 2013; 5:210-214. [PMID: 23888190 PMCID: PMC3722418 DOI: 10.4330/wjc.v5.i7.210] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 05/19/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023] Open
Abstract
Several epidemiological studies have clearly shown that low plasma levels of high density lipoprotein cholesterol (HDL-C) represent a cardiovascular disease (CVD) risk factor. However, it is unclear if there is a causal association between HDL-C concentration and CVD. A recent study published in the Lancet, which performed two Mendelian randomization analyses, showed that increased HDL-C levels were not associated with a decreased risk of myocardial infarction. These findings, together with the termination of the niacin-based AIM-HIGH trial and the discontinuation of cholesteryl ester transfer protein inhibitor dalcetrapib, challenge the concept that raising of plasma HDL-C will uniformly translate into reductions in CVD risk. HDL particles exhibit several anti-atherosclerotic properties, such as anti-inflammatory and anti-oxidative activities and cellular cholesterol efflux activity. Furthermore, HDL particles are very heterogeneous in terms of size, structure, composition and metabolism. HDL functionality may be associated more strongly with CVD risk than the traditional HDL-C levels. More research is needed to assess the association of the structure of HDL particle with its functionality and metabolism.
Collapse
|
24
|
Chang CT, Liao HY, Chang CM, Chen CY, Chen CH, Yang CY, Tsai FJ, Chen CJ. Oxidized ApoC1 on MALDI-TOF and glycated-ApoA1 band on gradient gel as potential diagnostic tools for atherosclerotic vascular disease. Clin Chim Acta 2013; 420:69-75. [DOI: 10.1016/j.cca.2012.10.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 10/09/2012] [Indexed: 12/20/2022]
|
25
|
NCAM function in the adult brain: lessons from mimetic peptides and therapeutic potential. Neurochem Res 2013; 38:1163-73. [PMID: 23494903 DOI: 10.1007/s11064-013-1007-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 02/01/2013] [Accepted: 02/18/2013] [Indexed: 01/22/2023]
Abstract
Neural cell adhesion molecules (NCAMs) are complexes of transmembranal proteins critical for cell-cell interactions. Initially recognized as key players in the orchestration of developmental processes involving cell migration, cell survival, axon guidance, and synaptic targeting, they have been shown to retain these functions in the mature adult brain, in relation to plastic processes and cognitive abilities. NCAMs are able to interact among themselves (homophilic binding) as well as with other molecules (heterophilic binding). Furthermore, they are the sole molecule of the central nervous system undergoing polysialylation. Most interestingly polysialylated and non-polysialylated NCAMs display opposite properties. The precise contributions each of these characteristics brings in the regulations of synaptic and cellular plasticity in relation to cognitive processes in the adult brain are not yet fully understood. With the aim of deciphering the specific involvement of each interaction, recent developments led to the generation of NCAM mimetic peptides that recapitulate identified binding properties of NCAM. The present review focuses on the information such advances have provided in the understanding of NCAM contribution to cognitive function.
Collapse
|
26
|
Wang S, Peng D. Regulation of adipocyte autophagy--the potential anti-obesity mechanism of high density lipoprotein and ApolipoproteinA-I. Lipids Health Dis 2012; 11:131. [PMID: 23039759 PMCID: PMC3478219 DOI: 10.1186/1476-511x-11-131] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 09/30/2012] [Indexed: 01/01/2023] Open
Abstract
Obesity is reaching epidemic worldwide and is risk factor for cardiovascular disease and type 2 diabetes. Although plasma high density lipoprotein (HDL) and apolipoprotein A-I (apoA-I) are inversely correlated to obesity, whether HDLs have anti-obesity effect remains unclear until a recent study reporting the direct anti-obesity effect of apoA-I and its mimetic peptide. However, the mechanism is not fully understood. Increasing adipose energy expenditure through attainment of brown adipocyte phenotype in white adipose tissue is considered a potential strategy to combat obesity. Specific inhibition of autophagy in adipose tissue is associated with reduced adiposity which is attributed to the attainment of brown adipocyte phenotype in white adipose tissue and the increased energy expenditure. HDL and apoA-I could activate PI3K-Akt-mTORC1 signaling which negatively regulates autophagy. The links between HDL/apoA-I and autophagy brings a new understanding on the anti-obesity effect of HDL and apoA-I.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, 139 Middle Ren-Min Rd, Changsha, Hunan 410011, China.
| | | |
Collapse
|