1
|
El‐Darzi N, Mast N, Li Y, Pikuleva IA. Dietary effects on the retina of hamsters. FASEB J 2025; 39:e70451. [PMID: 40099968 PMCID: PMC11917192 DOI: 10.1096/fj.202403390r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
The retina is a sensory tissue in the back of the eye, which captures visual information and relays it to the brain. The retinal pigment epithelium separates the neural retina from the choroidal (systemic) circulation and is thereby exposed to circulating lipoprotein particles. Herein, we used hamsters and conducted various retinal evaluations of animals fed either a normal diet or a Western-type diet (WTD). Prior to evaluations, hamsters were injected with indocyanine green (ICG), a fluorescent dye that binds to various proteins and lipids in the systemic circulation. The WTD increased plasma levels of total and HDL cholesterol 1.8- and 2.1-fold, respectively, and led to additional HDL2 and HDL3 subpopulations. The diet also increased the ICG fluorescence in the retinal pigment epithelium and the underlying choroidal circulation on histological tracking and altered retinal protein abundance as assessed by proteomics. Functional enrichments were found in the retinal gene expression, energy production, intracellular transport, cytoskeleton- and synapse-related processes, and protein ubiquitination. The biochemical basis linking the WTD, retinal energy production, and retinal neurotransmission was suggested as well. The data obtained were then compared with those from our previous investigations of hamsters and different mouse genotypes. We identified common retinal processes that can be affected by circulating lipoprotein particles regardless of the mechanism by which their levels and subpopulations were altered (through diet or genetic modification). Thus, we obtained novel mechanistic insights into how lipids in the systemic circulation can affect the retina.
Collapse
Affiliation(s)
- Nicole El‐Darzi
- Department of Ophthalmology and Visual SciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Natalia Mast
- Department of Ophthalmology and Visual SciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Yong Li
- Department of Ophthalmology and Visual SciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Irina A. Pikuleva
- Department of Ophthalmology and Visual SciencesCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
2
|
Jiang Q, Feng B, Zhao Y, Ren X, Dong C, Zhou R, Yang S, Gan W. Proatherogenic changes in the quantity and quality of lipoproteins in adults with idiopathic nephrotic syndrome. Clin Chim Acta 2025; 570:120206. [PMID: 39978456 DOI: 10.1016/j.cca.2025.120206] [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: 05/13/2024] [Revised: 01/01/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE Lipoprotein subclasses and high-density lipoprotein (HDL) functions are associated with atherosclerotic cardiovascular disease (ASCVD), but researches on them in patients with nephrotic syndrome (NS) are limited. The aims of this study were (1) to analyze the changes in quantity and quality of lipoprotein in patients with idiopathic nephrotic syndrome (INS) and patients in remission from NS, and (2) to evaluate the lipid-related atherosclerotic risk in these patients. METHODS 51 patients with idiopathic nephrotic syndrome (NS group), 72 NS patients with complete remission (NS remission group), and 80 healthy controls (control group) were recruited. The levels of conventional lipids, lipoprotein subclasses, including VLDL, IDL (C, B, A), LDL (LDL1-7), HDL (large, intermediate, small) and HDL cholesterol efflux capacity (CEC), were measured and compared across the three groups. RESULTS Conventional lipid parameters [TG, TC, LDL-C, apo-B and Lp(a)] and lipoprotein subclasses (VLDL, IDL-C, IDL-B, LDL-2 and sdLDL) were higher in NS group when compared to NS remission group and control group (P < 0.05). CEC in NS group was significantly lower than that in control group [21.0 (18.3-27.2) % vs 25.7 (23.3-28.9) %] (P < 0.001) and improved to 22.8 (20.6-23.7) % in NS remission group with the disease recovery. CONCLUSION Proatherogenic changes in conventional lipid parameters, lipoprotein subclasses and HDL-CEC were observed in patients with NS, suggesting that more rigorous lipid regulation strategies may help reduce cardiovascular disease risk in patients with NS.
Collapse
Affiliation(s)
- Qiuxiao Jiang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Clinical Laboratory, The First People's Hospital of Longquanyi District, Chengdu/West China (Longquan) Hospital, Sichuan University, Chengdu 610100 Sichuan, China
| | - Bin Feng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanhua Zhao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiangyi Ren
- Public Experimental Technology Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunmei Dong
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruyi Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuyu Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Gan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Zhu L, Li L, Wu J. FcRn inhibitors: Transformative advances and significant impacts on IgG-mediated autoimmune diseases. Autoimmun Rev 2025; 24:103719. [PMID: 39672251 DOI: 10.1016/j.autrev.2024.103719] [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: 09/10/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Pathogenic IgG autoantibodies play a crucial role in the pathogenesis of autoimmune diseases, and removal of pathogenic IgG autoantibodies is an important therapeutic approach and tool for such diseases. The neonatal Fc receptor (FcRn) interacts with IgG and protects it from lysosomal degradation. FcRn inhibitors accelerate the clearance of IgG antibodies, including pathogenic IgG autoantibodies, by targeting and blocking the binding of FcRn to IgG. Theoretically, FcRn inhibitors can be applied for the treatment of IgG-mediated autoimmune diseases. With successful completion of multiple relevant clinical trials, key evidence-based data have been provided for FcRn inhibitors in the treatment of IgG-mediated autoimmune diseases, and several FcRn inhibitors have been approved for these indications. Additional trials are being planned or conducted. This review examines all available high-quality clinical trials of FcRn inhibitors assessing IgG-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Lina Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China
| | - Lanjun Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China..
| |
Collapse
|
4
|
Antozzi C, Vu T, Ramchandren S, Nowak RJ, Farmakidis C, Bril V, De Bleecker J, Yang H, Minks E, Park JS, Grudniak M, Smilowski M, Sevilla T, Hoffmann S, Sivakumar K, Suzuki Y, Youssef E, Sanga P, Karcher K, Zhu Y, Sheehan JJ, Sun H. Safety and efficacy of nipocalimab in adults with generalised myasthenia gravis (Vivacity-MG3): a phase 3, randomised, double-blind, placebo-controlled study. Lancet Neurol 2025; 24:105-116. [PMID: 39862879 DOI: 10.1016/s1474-4422(24)00498-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/14/2024] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Given burdensome side-effects and long latency for efficacy with conventional agents, there is a continued need for generalised myasthenia gravis treatments that are safe and provide consistently sustained, long-term disease control. Nipocalimab, a neonatal Fc receptor blocker, was associated with dose-dependent reductions in total IgG and anti-acetylcholine receptor (AChR) antibodies and clinically meaningful improvements in the Myasthenia Gravis Activities of Daily Living (MG-ADL) scale in patients with generalised myasthenia gravis in a phase 2 study. We aimed to assess the safety and efficacy of nipocalimab in a phase 3 study. METHODS Vivacity-MG3 was a phase 3, randomised, double-blind, placebo-controlled, phase 3 study conducted at 81 outpatient centres with expertise in myasthenia gravis in 17 countries in Asia-Pacific, Europe, and North America. Adults (aged ≥18 years) with generalised myasthenia gravis inadequately controlled with standard-of-care therapy (MG-ADL score ≥6) were randomly assigned (1:1) to either nipocalimab (30 mg/kg loading dose then 15 mg/kg every 2 weeks for maintenance dosing) or placebo infusions every 2 weeks, added to standard-of-care therapy in both groups, for 24 weeks. Randomisation was stratified by antibody status, day 1 MG-ADL total score, and region. The sponsor, investigators, clinical raters, and participants were masked to treatment assignment. The primary endpoint was the difference between nipocalimab and placebo based on least-squares mean change from baseline in MG-ADL total score averaged over weeks 22, 23, and 24 in the intention-to-treat population of patients who were antibody-positive (for AChR, anti-muscle-specific tyrosine kinase [MuSK], or anti-low-density lipoprotein receptor-related protein 4 [LRP4]). Adverse events were assessed in patients who received at least one dose of study drug. This study is registered at ClinicalTrials.gov, NCT04951622; the double-blind phase is completed and an open-label extension phase is ongoing. FINDINGS Between July 15, 2021, and Nov 17, 2023, 199 patients were enrolled, and 196 patients received study drug (98 in the nipocalimab group and 98 in the placebo group); of these, 153 (77 in the nipocalimab group and 76 in the placebo group) were antibody-positive. The least-squares mean change in MG-ADL score from baseline to weeks 22, 23, and 24 was -4·70 (SE 0·329) in the nipocalimab group versus -3·25 (0·335) in the placebo group (difference -1·45 [95% CI -2·38 to -0·52]; p=0·0024). The incidence of adverse events was similar between groups (82 [84%] of 98 in both the nipocalimab and placebo groups), including infections (42 [43%] of 98 in the nipocalimab group and placebo group) and headache (14 [14%] of 98 in the nipocalimab group and 17 [17%] of 98 in the placebo group). Serious adverse events were reported for nine (9%) of 98 patients in the nipocalimab group and 14 (14%) of 98 patients in the placebo group, three of which had a fatal outcome (nipocalimab: myasthenic crisis; placebo: cardiac arrest and myocardial infarction). INTERPRETATION Results from the completed double-blind phase of Vivacity-MG3 support the role of nipocalimab, added to standard-of-care therapies, as a safe treatment for sustained disease control over 6 months for a broad population of patients with generalised myasthenia gravis who are antibody-positive. The ongoing open-label extension phase should provide longer term sustained safety and efficacy data with nipocalimab. FUNDING Janssen Research & Development, LLC, a Johnson & Johnson company.
Collapse
Affiliation(s)
- Carlo Antozzi
- Neuroimmunology and Neuromuscular Diseases Unit, and Apheresis and Immunotherapy Unit, IRCCS Carlo Besta Neurological Institute Foundation, Milan, Italy
| | - Tuan Vu
- Department of Neurology, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sindhu Ramchandren
- Janssen Research & Development, a Johnson & Johnson Company, Titusville, NJ, USA.
| | - Richard J Nowak
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Vera Bril
- Department of Medicine, University of Toronto, University Health Network, Toronto, ON, Canada
| | - Jan De Bleecker
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, China
| | - Eduard Minks
- Department of Neurology, Masaryk University and St Anne's Hospital, Brno, Czechia
| | - Jin-Sung Park
- Department of Neurology, School of Medicine, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | | | | | - Teresa Sevilla
- Department of Medicine, Hospital Universitari i Politècnic and IIS La Fe and University of Valencia, Valencia, Spain
| | - Sarah Hoffmann
- Department of Neurology, Neuroscience Clinical Research Center (NCRC) and Integrated Myasthenia Gravis Centre, Charité Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kumaraswamy Sivakumar
- The Neuromuscular Research Centre and Neuromuscular Clinic of Arizona, Phoenix, AZ, USA
| | - Yasushi Suzuki
- Department of Neurology, National Hospital Organisation Sendai Medical Centre, Sendai, Japan
| | - Eriene Youssef
- Janssen Research & Development, a Johnson & Johnson Company, Titusville, NJ, USA
| | - Panna Sanga
- Janssen Research & Development, a Johnson & Johnson Company, Titusville, NJ, USA
| | - Keith Karcher
- Statistics and Decision Sciences, Janssen Research & Development, a Johnson & Johnson Company, Titusville, NJ, USA
| | - Yaowei Zhu
- Janssen Research & Development, a Johnson & Johnson Company, Titusville, NJ, USA
| | - John J Sheehan
- Global Medical Affairs, Janssen Global Services, a Johnson & Johnson Company, Raritan, NJ, USA
| | - Hong Sun
- Janssen Research & Development, a Johnson & Johnson Company, Titusville, NJ, USA
| |
Collapse
|
5
|
Lehmann M, Halder S, Reinholdt P, Bashawat M, Scheidt HA, Leopold J, Schiller J, di Prima D, Akkerman V, Szomek M, Lauritsen L, Kongsted J, Müller P, Wessig P, Wüstner D. Synthesis and Characterization of a Novel Intrinsically Fluorescent Analog of Cholesterol with Improved Photophysical Properties. Anal Chem 2024; 96:18596-18604. [PMID: 39537343 PMCID: PMC11603404 DOI: 10.1021/acs.analchem.3c05720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Live-cell imaging of cholesterol trafficking depends on suitable cholesterol analogs. However, existing fluorescent analogs of cholesterol either show very different physicochemical properties compared to cholesterol or demand excitation in the ultraviolet spectral region. We present a strategy to synthesize two novel intrinsically fluorescent sterol probes with a close resemblance of cholesterol. The analogs contain four conjugated double bonds in the ring system and either a keto group (probe 5) or a hydroxy group (probe 6) in the C3 position. The emission of 5 is in the visible range of the spectrum, i.e., red-shifted by 150 nm compared to the widely used dehydroergosterol. Together with its high multiphoton absorption, this allows for imaging of 5 on conventional microscopes, including multicolor 3D and time-lapse microscopy. Molecular dynamics simulations and nuclear magnetic resonance spectroscopy reveal that 5 can condense the fatty acyl chains of phospholipids in model membranes. In giant unilamellar vesicles, 5 partitions equally into the liquid-ordered and disordered phases. In contrast, 6 emits in the ultraviolet range and is unstable in solution, preventing its use in live-cell imaging applications. The good photophysical properties of 5 make it a suitable analogue for improved live-cell imaging of sterol transport.
Collapse
Affiliation(s)
- Max Lehmann
- Department
of Chemistry, University of Potsdam, Karl-Liebknecht Str. 24-25, Potsdam 14476, Germany
| | - Senjuti Halder
- Department
of Biochemistry and Molecular Biology, University
of Southern Denmark, Odense
M DK-5230, Denmark
| | - Peter Reinholdt
- Department
of Physics, Chemistry and Pharmacy, University
of Southern Denmark, Odense M DK-5230, Denmark
| | - Mohammad Bashawat
- Department
of Biology, Humboldt University Berlin, Invalidenstr. 42, Berlin 10115, Germany
| | - Holger A. Scheidt
- Institute
for Medical Physics and Biophysics, Leipzig
University, Härtelstr.
16-18, Leipzig 04107, Germany
| | - Jenny Leopold
- Institute
for Medical Physics and Biophysics, Leipzig
University, Härtelstr.
16-18, Leipzig 04107, Germany
| | - Jürgen Schiller
- Institute
for Medical Physics and Biophysics, Leipzig
University, Härtelstr.
16-18, Leipzig 04107, Germany
| | - Duccio di Prima
- Department
of Physics, Chemistry and Pharmacy, University
of Southern Denmark, Odense M DK-5230, Denmark
| | - Vibeke Akkerman
- Department
of Biochemistry and Molecular Biology, University
of Southern Denmark, Odense
M DK-5230, Denmark
| | - Maria Szomek
- Department
of Biochemistry and Molecular Biology, University
of Southern Denmark, Odense
M DK-5230, Denmark
| | - Line Lauritsen
- Department
of Biochemistry and Molecular Biology, University
of Southern Denmark, Odense
M DK-5230, Denmark
| | - Jacob Kongsted
- Department
of Physics, Chemistry and Pharmacy, University
of Southern Denmark, Odense M DK-5230, Denmark
| | - Peter Müller
- Department
of Biology, Humboldt University Berlin, Invalidenstr. 42, Berlin 10115, Germany
| | - Pablo Wessig
- Department
of Chemistry, University of Potsdam, Karl-Liebknecht Str. 24-25, Potsdam 14476, Germany
| | - Daniel Wüstner
- Department
of Biochemistry and Molecular Biology, University
of Southern Denmark, Odense
M DK-5230, Denmark
| |
Collapse
|
6
|
Chien SC, Lan WR, Yun CH, Sung KT, Yeh HI, Tsai CT, Chien CY, Hung CL. Functional and prognostic impacts of serum albumin with thoracic arterial calcification among asymptomatic individuals. Sci Rep 2024; 14:25477. [PMID: 39462060 PMCID: PMC11513936 DOI: 10.1038/s41598-024-77228-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
Although several studies have demonstrated the cardiovascular (CV) implication of hypoalbuminemia and arterial calcification among hemodialysis patients, little is known regarding their cardiac correlates and relevant CV outcomes in asymptomatic individuals. We assessed the potential CV interrelation between serum albumin (SA) and aortic calcification. Among 2,723 asymptomatic individuals underwent cardiovascular health check-up, we assessed serum albumin (SA) level, thoracic aortic calcification (TAC) and coronary artery calcification (CAC) by multi-detector computed tomography, and ultrasound-determined carotid plaque burden. We related these measures to cardiac structure/function and CV outcomes. Lower SA level was associated with higher TAC score and volume rather than carotid plaque or coronary calcification burden in fully adjusted models. By categorizing the study population into 4 groups by SA (>, ≤ 4.6 mg/dL) and presence of TAC, subjects classified into low SA/TAC(+) category were oldest with highest prevalent CV disease. Both lower SA and TAC(+) were independently associated with impaired myocardial systolic/diastolic mechanics and higher CV events during a median of 6.6 years (IQR: 5.1, 6.8 years) follow-up. Participants classified into low SA/TAC(+) category showed highest risk for CV events (adjusted HR: 3.78 [95% CI: 2.11, 6.77], high SA/TAC[-] as reference) in fully adjusted Cox model. Among symptom-free individuals, TAC was closely associated with low SA concentration in relation to unfavorable cardiac mechanics and may serve as a useful prognosticator for adverse CV events.
Collapse
Grants
- NSC-101-2314-B-195-020, NSC103-2314-B-010-005-MY3, 103-2314-B-195-001-MY3, 101-2314-B-195-020-MY1, MOST 103-2314-B-195-006-MY3, NSC102-2314-B-002-046-MY3, 106-2314-B-195-008-MY2, 108-2314-B-195-018-MY2, MOST 108-2314-B-195-018-MY2, MOST 109-2314-B-715-008, and MOST 110-2314-B-715-009-MY1 Ministry of Science and Technology, Taiwan
- 10271, 10248, 10220, 10253, 10375, 10358, and E-102003 Mackay Memorial Hospital
- Taiwan Foundation for geriatric emergency and critical care
Collapse
Affiliation(s)
- Shih-Chieh Chien
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan
| | - Wei-Ren Lan
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan
| | - Chun-Ho Yun
- Department of Radiology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Tzu Sung
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan
| | - Hung-I Yeh
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan
| | - Cheng-Ting Tsai
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan
| | - Chen-Yen Chien
- Division of Cardiovascular Surgery, Department of Surgery, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan.
| | - Chung-Lieh Hung
- Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, No. 92, Sec. 2, Zhongshan N. Rd.,, Taipei, 10449, Taiwan.
| |
Collapse
|
7
|
Lorenz J, Eisenhardt C, Mittermair T, Kulle AE, Holterhus PM, Fobker M, Boenigk W, Nordhoff V, Behre HM, Strünker T, Brenker C. The sperm-specific K + channel Slo3 is inhibited by albumin and steroids contained in reproductive fluids. Front Cell Dev Biol 2024; 12:1275116. [PMID: 39310227 PMCID: PMC11413451 DOI: 10.3389/fcell.2024.1275116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 07/19/2024] [Indexed: 09/25/2024] Open
Abstract
To locate and fertilize the egg, sperm probe the varying microenvironment prevailing at different stages during their journey across the female genital tract. To this end, they are equipped with a unique repertoire of mostly sperm-specific proteins. In particular, the flagellar Ca2+ channel CatSper has come into focus as a polymodal sensor used by human sperm to register ligands released into the female genital tract. Here, we provide the first comprehensive study on the pharmacology of the sperm-specific human Slo3 channel, shedding light on its modulation by reproductive fluids and their constituents. We show that seminal fluid and contained prostaglandins and Zn2+ do not affect the channel, whereas human Slo3 is inhibited in a non-genomic fashion by diverse steroids as well as by albumin, which are released into the oviduct along with the egg. This indicates that not only CatSper but also Slo3 harbours promiscuous ligand-binding sites that can accommodate structurally diverse molecules, suggesting that Slo3 is involved in chemosensory signalling in human sperm.
Collapse
Affiliation(s)
- Johannes Lorenz
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Clara Eisenhardt
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Teresa Mittermair
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Alexandra E. Kulle
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Christian-Albrechts-University, Kiel, Germany
| | - Paul Martin Holterhus
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Christian-Albrechts-University, Kiel, Germany
| | - Manfred Fobker
- Center for Laboratory Medicine, University Hospital, Münster, Germany
| | - Wolfgang Boenigk
- Max Planck Institute for Neurobiology of Behaviour—Caesar, Bonn, Germany
| | - Verena Nordhoff
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | | | - Timo Strünker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| |
Collapse
|
8
|
Ma G, Crowley AR, Heyndrickx L, Rogiers I, Parthoens E, Van Santbergen J, Ober RJ, Bobkov V, de Haard H, Ulrichts P, Hofman E, Louagie E, Balbino B, Ward ES. Differential effects of FcRn antagonists on the subcellular trafficking of FcRn and albumin. JCI Insight 2024; 9:e176166. [PMID: 38713534 PMCID: PMC11141909 DOI: 10.1172/jci.insight.176166] [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: 10/04/2023] [Accepted: 04/10/2024] [Indexed: 05/09/2024] Open
Abstract
The homeostasis of IgG is maintained by the neonatal Fc receptor, FcRn. Consequently, antagonism of FcRn to reduce endogenous IgG levels is an emerging strategy for treating antibody-mediated autoimmune disorders using either FcRn-specific antibodies or an engineered Fc fragment. For certain FcRn-specific antibodies, this approach has resulted in reductions in the levels of serum albumin, the other major ligand transported by FcRn. Cellular and molecular analyses of a panel of FcRn antagonists have been carried out to elucidate the mechanisms leading to their differential effects on albumin homeostasis. These analyses have identified 2 processes underlying decreases in albumin levels during FcRn blockade: increased degradation of FcRn and competition between antagonist and albumin for FcRn binding. These findings have potential implications for the design of drugs to modulate FcRn function.
Collapse
Affiliation(s)
- Guanglong Ma
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew R. Crowley
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | - Eef Parthoens
- VIB BioImaging Core, Center for Inflammation Research, Ghent, Belgium
| | | | - Raimund J. Ober
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | | | | | | | | | - E. Sally Ward
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
9
|
Antozzi C, Guptill J, Bril V, Gamez J, Meuth SG, Nowak RJ, Quan D, Sevilla T, Jouvin MH, Jin J, Karcher K, Ramchandren S, Sun H, Ling L, Zhu Y, Arroyo S. Safety and Efficacy of Nipocalimab in Patients With Generalized Myasthenia Gravis: Results From the Randomized Phase 2 Vivacity-MG Study. Neurology 2024; 102:e207937. [PMID: 38165333 PMCID: PMC10962909 DOI: 10.1212/wnl.0000000000207937] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/20/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES To evaluate in a phase 2 study the safety and efficacy of IV nipocalimab, a fully human, antineonatal Fc receptor monoclonal antibody, in patients with generalized myasthenia gravis (gMG). METHODS Patients with gMG with inadequate response to stable standard-of-care (SOC) therapy were randomized 1:1:1:1:1 to receive either IV placebo every 2 weeks (Q2W) or one of 4 IV nipocalimab treatments: 5 mg/kg once every 4 weeks (Q4W), 30 mg/kg Q4W, 60 mg/kg Q2W each for 8 weeks, or a 60 mg/kg single dose, in addition to their background SOC therapy. Infusions (placebo or nipocalimab) were Q2W in all groups to maintain blinding. The primary safety endpoint was incidence of treatment-emergent adverse events (TEAEs), including serious adverse events and adverse events of special interest. The primary efficacy endpoint was change from baseline to day 57 in Myasthenia Gravis-Activities of Daily Living (MG-ADL) total scores. Dose response of change at day 57 was analyzed with a linear trend test over the placebo, nipocalimab 5 mg/kg Q4W, nipocalimab 30 mg/kg Q4W, and nipocalimab 60 mg/kg Q2W groups. RESULTS Sixty-eight patients (nipocalimab: n = 54; placebo, n = 14) were randomized; 64 patients (94.1%) were positive for antiacetylcholine receptor autoantibodies, and 4 patients (6%) were positive for antimuscle-specific tyrosine kinase autoantibodies. Fifty-seven patients (83.8%) completed treatment through day 57. The combined nipocalimab group compared with the placebo group demonstrated similar incidences of TEAEs (83.3% vs 78.6%, respectively) and infections (33.3% vs 21.4%, respectively). No deaths or discontinuations due to TEAEs and no TEAEs of special interest (grade ≥3 infection or hypoalbuminemia) were observed with nipocalimab treatment. A statistically significant dose response was observed for change from baseline in MG-ADL at day 57 (p = 0.031, test of linear trend). DISCUSSION Nipocalimab was generally safe, well-tolerated, and showed evidence of dose-dependent reduction in MG-ADL scores at day 57 in this phase 2 study. These results support further evaluation of nipocalimab for the treatment of gMG. TRIAL REGISTRATION INFORMATION Clinical Trials Registration: NCT03772587; first submitted December 10, 2018; EudraCT Number: 2018-002247-28; first submitted November 30, 2018; date of first patient dosed April 10, 2019. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that for patients with gMG, nipocalimab was well-tolerated, and it did not significantly improve MG-ADL at any individual dose but demonstrated a significant dose response for improved MG-ADL across doses.
Collapse
Affiliation(s)
- Carlo Antozzi
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Jeffrey Guptill
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Vera Bril
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Josep Gamez
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Sven G Meuth
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Richard J Nowak
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Dianna Quan
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Teresa Sevilla
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Marie-Helene Jouvin
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Jim Jin
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Keith Karcher
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Sindhu Ramchandren
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Hong Sun
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Leona Ling
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Yaowei Zhu
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| | - Santiago Arroyo
- From the Neurological Institute Foundation C. Besta (C.A.), Milan, Italy; Duke University School of Medicine (J. Guptill), Durham, NC; Argenx US Inc. (J. Guptill), Boston, MA; University of Toronto (V.B.), ON, Canada; Universitat Autonoma de Barcelona, (J. Gamez), Spain; Medical Faculty (S.G.M.), Heinrich-Heine-University, Düsseldorf, Germany; Yale University School of Medicine (R.J.N.), New Haven, CT; University of Colorado School of Medicine (D.Q.), Aurora; Hospital Universitari i Politécnic La Fe (T.S.), Universitat de Valencia, Spain; Pharvaris, Inc. (M.-H.J.), Boston, MA; Janssen Research & Development, LLC, (J.J., K.K., S.R.,H.S., L.L., Y.Z.), Titusville, NJ; Marinus Pharmaceuticals, Inc. Radnor, PA; Fulcrum Therapeutics (S.A.), Cambridge, MA
| |
Collapse
|
10
|
Minaychev VV, Teterina AY, Smirnova PV, Menshikh KA, Senotov AS, Kobyakova MI, Smirnov IV, Pyatina KV, Krasnov KS, Fadeev RS, Komlev VS, Fadeeva IS. Composite Remineralization of Bone-Collagen Matrices by Low-Temperature Ceramics and Serum Albumin: A New Approach to the Creation of Highly Effective Osteoplastic Materials. J Funct Biomater 2024; 15:27. [PMID: 38391880 PMCID: PMC10889756 DOI: 10.3390/jfb15020027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
This study examined the effectiveness of coating demineralized bone matrix (DBM) with amorphous calcium phosphate (DBM + CaP), as well as a composite of DBM, calcium phosphate, and serum albumin (DBM + CaP + BSA). The intact structure of DBM promotes the transformation of amorphous calcium phosphate (CaP) into dicalcium phosphate dihydrate (DCPD) with a characteristic plate shape and particle size of 5-35 µm. The inclusion of BSA in the coating resulted in a better and more uniform distribution of CaP on the surface of DBM trabeculae. MG63 cells showed that both the obtained forms of CaP and its complex with BSA did not exhibit cytotoxicity up to a concentration of 10 mg/mL in vitro. Ectopic (subcutaneous) implantation in rats revealed pronounced biocompatibility, as well as strong osteoconductive, osteoinductive, and osteogenic effects for both DBM + CaP and DBM + CaP + BSA, but more pronounced effects for DBM + CaP + BSA. In addition, for the DBM + CaP + BSA samples, there was a pronounced full physiological intrafibrillar biomineralization and proangiogenic effect with the formation of bone-morrow-like niches, accompanied by pronounced processes of intramedullary hematopoiesis, indicating a powerful osteogenic effect of this composite.
Collapse
Affiliation(s)
- Vladislav V Minaychev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Anastasia Yu Teterina
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Polina V Smirnova
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Ksenia A Menshikh
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Anatoliy S Senotov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Margarita I Kobyakova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 630060 Novosibirsk, Russia
| | - Igor V Smirnov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Kira V Pyatina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Kirill S Krasnov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Roman S Fadeev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Vladimir S Komlev
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Irina S Fadeeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| |
Collapse
|
11
|
El-Darzi N, Mast N, Li Y, Pikuleva IA. APOB100 transgenic mice exemplify how the systemic circulation content may affect the retina without altering retinal cholesterol input. Cell Mol Life Sci 2024; 81:52. [PMID: 38253888 PMCID: PMC10803575 DOI: 10.1007/s00018-023-05056-4] [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: 09/08/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 01/24/2024]
Abstract
Apolipoprotein B (APOB) is a constituent of unique lipoprotein particles (LPPs) produced in the retinal pigment epithelium (RPE), which separates the neural retina from Bruch's membrane (BrM) and choroidal circulation. These LPPs accumulate with age in BrM and contribute to the development of age-related macular degeneration, a major blinding disease. The APOB100 transgenic expression in mice, which unlike humans lack the full-length APOB100, leads to lipid deposits in BrM. Herein, we further characterized APOB100 transgenic mice. We imaged mouse retina in vivo and assessed chorioretinal lipid distribution, retinal sterol levels, retinal cholesterol input, and serum content as well as tracked indocyanine green-bound LPPs in mouse plasma and retina after an intraperitoneal injection. Retinal function and differentially expressed proteins were also investigated. APOB100 transgenic mice had increased serum LDL content and an additional higher density HDL subpopulation; their retinal cholesterol levels (initially decreased) became normal with age. The LPP cycling between the RPE and choroidal circulation was increased. Yet, LPP trafficking from the RPE to the neural retina was limited, and total retinal cholesterol input did not change. There were lipid deposits in the RPE and BrM, and retinal function was impaired. Retinal proteomics provided mechanistic insights. Collectively, our data suggested that the serum LDL/HDL ratio may not affect retinal pathways of cholesterol input as serum LPP load is mainly handled by the RPE, which offloads LPP excess to the choroidal circulation rather than neural retina. Different HDL subpopulations should be considered in studies linking serum LPPs and age-related macular degeneration.
Collapse
Affiliation(s)
- Nicole El-Darzi
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yong Li
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
12
|
King TE, Humphrey JR, Laughton CA, Thomas NR, Hirst JD. Optimizing Excipient Properties to Prevent Aggregation in Biopharmaceutical Formulations. J Chem Inf Model 2024; 64:265-275. [PMID: 38113509 PMCID: PMC10777730 DOI: 10.1021/acs.jcim.3c01898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Excipients are included within protein biotherapeutic solution formulations to improve colloidal and conformational stability but are generally not designed for the specific purpose of preventing aggregation and improving cryoprotection in solution. In this work, we have explored the relationship between the structure and antiaggregation activity of excipients by utilizing coarse-grained molecular dynamics modeling of protein-excipient interaction. We have studied human serum albumin as a model protein, and we report the interaction of 41 excipients (polysorbates, fatty alcohol ethoxylates, fatty acid ethoxylates, phospholipids, glucosides, amino acids, and others) in terms of the reduction of solvent accessible surface area of aggregation-prone regions, proposed as a mechanism of aggregation prevention. Polyoxyethylene sorbitan had the greatest degree of interaction with aggregation-prone regions, decreasing the solvent accessible surface area of APRs by 20.7 nm2 (40.1%). Physicochemical descriptors generated by Mordred are employed to probe the structure-property relationship using partial least-squares regression. A leave-one-out cross-validated model had a root-mean-square error of prediction of 4.1 nm2 and a mean relative error of prediction of 0.077. Generally, longer molecules with a large number of alcohol-terminated PEG units tended to interact more, with qualitatively different protein interactions, wrapping around the protein. Shorter or less ethoxylated compounds tend to form hemimicellar clusters at the protein surface. We propose that an improved design would feature many short chains of 5 to 10 PEG units in many distinct branches and at least some hydrophobic content in the form of medium-length or greater aliphatic chains (i.e., six or more carbon atoms). The combination of molecular dynamics simulation and quantitative modeling is an important first step in an all-purpose protein-independent model for the computer-aided design of stabilizing excipients.
Collapse
Affiliation(s)
- Toby E. King
- Biodiscovery
Institute, School of Pharmacy, University Park, Nottingham NG7 2RD, U.K.
| | | | - Charles A. Laughton
- Biodiscovery
Institute, School of Pharmacy, University Park, Nottingham NG7 2RD, U.K.
| | - Neil R. Thomas
- Biodiscovery
Institute, School of Chemistry, University Park, Nottingham NG7 2RD, U.K.
| | | |
Collapse
|
13
|
Schachtl-Riess JF, Schönherr S, Lamina C, Forer L, Coassin S, Streiter G, Kheirkhah A, Li Y, Meiselbach H, Di Maio S, Eckardt KU, Köttgen A, Kronenberg F. KLKB1 and CLSTN2 are associated with HDL-mediated cholesterol efflux capacity in a genome-wide association study. Atherosclerosis 2023; 368:1-11. [PMID: 36812656 DOI: 10.1016/j.atherosclerosis.2023.01.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/06/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS HDL-mediated cholesterol efflux capacity (CEC) may protect from cardiovascular disease. Thus, we aimed to identify its genetic and non-genetic determinants. METHODS We measured CEC to 2% apolipoprotein B-depleted serum using BODIPY-cholesterol and cAMP-stimulated J774A.1 macrophages using serum samples from 4,981 participants in the German Chronic Kidney Disease (GCKD) study. Variance of CEC explained by clinical and biochemical parameters in a multivariable linear regression model was calculated by proportional marginal variance decomposition. A genome-wide association study with 7,746,917 variants was performed based on an additive genetic model. The main model was adjusted for age, sex and principal components 1-10. Further models were selected for sensitivity analysis and to reduce residual variance by known CEC pathways. RESULTS Variables that explained 1% and more of the variance of CEC were concentrations of triglycerides (12.9%), HDL-cholesterol (11.8%), LDL-cholesterol (3.0%), apolipoprotein A-IV (2.8%), PCSK9 (1.0%), and eGFR (1.0%). The KLKB1 (chr4) and APOE/C1 (chr19) loci were genome-wide significantly (p < 5x10-8) associated with CEC in our main model (p = 8.8x10-10 and p = 3.3x10-10, respectively). KLKB1 remained significantly associated after additional adjustment for either kidney parameters, HDL-cholesterol, triglycerides or apolipoprotein A-IV concentrations, while the APOE/C1 locus was not significantly associated anymore after adjustment for triglycerides. Adjustment for triglycerides also revealed an association with the CLSTN2 locus (chr3; p = 6.0x10-9). CONCLUSIONS We identified HDL-cholesterol and triglycerides as the main determinants of CEC. Furthermore, we newly found a significant association of CEC with the KLKB1 and the CLSTN2 locus and confirmed the association with the APOE/C1 locus, likely mediated by triglycerides.
Collapse
Affiliation(s)
- Johanna F Schachtl-Riess
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gertraud Streiter
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silvia Di Maio
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
14
|
Development and validation of novel automatable assay for cholesterol efflux capacity. Biosci Rep 2023; 43:232455. [PMID: 36645426 PMCID: PMC9905788 DOI: 10.1042/bsr20221519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/24/2022] [Accepted: 01/16/2023] [Indexed: 01/17/2023] Open
Abstract
During the past decade, evaluation of high-density lipoprotein (HDL) functionality has been well studied for predicting cardiovascular disease (CVD) risk. Cholesterol efflux capacity (CEC) is the strongest candidate as the biomarker out of various HDL antiatherosclerotic functions. However, CEC has not yet been introduced clinically because of several technical issues, including the use of radioactive materials and differentiated cells in the assay. Previously, our laboratory developed a radioisotope- and cell-free CEC assay called the immobilized liposome-bound gel beads (ILGs) method to replace the conventional method. However, the separation process of the supernatant was not suitable for installation in an automatic analyzer. The present study aims to develop a new method that is easier to operate. We assumed that the use of magnetic beads instead of gel beads would enable the skip of the centrifugal process. First, similar to the ILG method, porous magnetic beads were treated with liposomes containing fluorescently labeled cholesterol. Fluorescence was observed inside the magnetic beads, and almost the same amount of liposomes as in the ILG method was immobilized successfully. These immobilized liposome-bound magnetic beads (ILMs) were available for CEC assay when HDL and apolipoprotein B-100-depleted serum (BDS) were used as cholesterol acceptors. The ILM method showed sufficient basic performance and a good correlation with the ILG method. Furthermore, when the CEC of 15 serum samples from healthy subjects was measured, a good correlation between HDL-cholesterol level and the ILG method was confirmed. Thus, it was confirmed that the ILM method was successfully developed and could be automated.
Collapse
|
15
|
de Rojas I, del Barrio L, Hernández I, Montrreal L, García-González P, Marquié M, Valero S, Cano A, Orellana A, Boada M, Mañes S, Ruiz A. Correlations between the NMR Lipoprotein Profile, APOE Genotype, and Cholesterol Efflux Capacity of Fasting Plasma from Cognitively Healthy Elderly Adults. Int J Mol Sci 2023; 24:ijms24032186. [PMID: 36768512 PMCID: PMC9916740 DOI: 10.3390/ijms24032186] [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: 11/17/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Cholesterol efflux capacity (CEC) is of interest given its potential relationship with several important clinical conditions including Alzheimer's disease. The inactivation of the APOE locus in mouse models supports the idea that it is involved in determining the CEC. With that in mind, we examine the impact of the plasma metabolome profile and the APOE genotype on the CEC in cognitively healthy elderly subjects. The study subjects were 144 unrelated healthy individuals. The plasma CEC was determined by exposing cultured mouse macrophages treated with BODIPY-cholesterol to human plasma. The metabolome profile was determined using NMR techniques. Multiple regression was performed to identify the most important predictors of CEC, as well as the NMR features most strongly associated with the APOE genotype. Plasma 3-hydroxybutyrate was the variable most strongly correlated with the CEC (r = 0.365; p = 7.3 × 10-6). Male sex was associated with a stronger CEC (r = -0.326, p = 6.8 × 10-5). Most of the NMR particles associated with the CEC did not correlate with the APOE genotype. The NMR metabolomics results confirmed the APOE genotype to have a huge effect on the concentration of plasma lipoprotein particles as well as those of other molecules including omega-3 fatty acids. In conclusion, the CEC of human plasma was associated with ketone body concentration, sex, and (to a lesser extent) the other features of the plasma lipoprotein profile. The APOE genotype exerted only a weak effect on the CEC via the modulation of the lipoprotein profile. The APOE locus was associated with omega-3 fatty acid levels independent of the plasma cholesterol level.
Collapse
Affiliation(s)
- Itziar de Rojas
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura del Barrio
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Isabel Hernández
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Montrreal
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
| | - Pablo García-González
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergi Valero
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amanda Cano
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain
- Correspondence: (S.M.); (A.R.)
| | - Agustín Ruiz
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (S.M.); (A.R.)
| |
Collapse
|
16
|
Chang CH, Yeh ST, Ooi SW, Li CY, Chen HF. The relationship of low-density lipoprotein cholesterol and all-cause or cardiovascular mortality in patients with type 2 diabetes: a retrospective study. PeerJ 2023; 11:e14609. [PMID: 36643628 PMCID: PMC9835695 DOI: 10.7717/peerj.14609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/30/2022] [Indexed: 01/11/2023] Open
Abstract
Background The optimal levels of low-density lipoprotein cholesterol (LDL-C) in patients with type 2 diabetes (T2D) are not currently clear. In this study, we determined the relationship between various mean LDL-C and all-cause or cardiovascular mortality risks in patients with T2D, stratifying by albumin level, age, sex, and antilipid medication use. We also evaluated the association of LDL-C standard deviation (LDL-C-SD) and all-cause and cardiovascular mortality by type of antilipid medication use. Methods A total of 46,675 T2D patients with a prescription for antidiabetic agents >6 months from outpatient visits (2003-2018) were linked to Taiwan's National Death Registry to identify all-cause and cardiovascular mortality. The Poisson assumption was used to estimate mortality rates, and the Cox proportional hazard regression model was used to assess the relative hazards of respective mortality in relation to mean LDL-C in patient cohorts by albumin level, age, sex, and antilipid use adjusting for medications, comorbidities, and laboratory results. We also determined the overall, and anti-lipid-specific mortality rates and relative hazards of all-cause and cardiovascular mortality associated with LDL-C-SD using the Poisson assumption and Cox proportional hazard regression model, respectively. Results All-cause and cardiovascular mortality rates were the lowest in T2D patients with a mean LDL-C > 90-103.59 mg/dL in the normal albumin group (≥ 3.5 g/dL). Compared to T2D patients with a mean LDL-C > 90-103.59 mg/dL, those with a mean LDL-C ≤ 77 mg/dL had an elevated risk of all-cause mortality in both the normal and lower albumin groups. T2D patients with a mean LDL-C ≤ 90 and > 103.59-119 mg/dL had relatively higher risk of cardiovascular mortality in the normal albumin group, but in the lower albumin group (<3.5 g/dL), any level of mean LDL-C ≤ 119 mg/dL was not significantly associated with cardiovascular mortality. Increased risks of all-cause and cardiovascular mortality were observed in patients with a mean LDL-C ≤ 77 mg/dL in both sexes and in all age groups except in those aged <50 years, a lower mean LDL-C was not associated with cardiovascular mortality. Similarly, patients with an LDL-C-SD <10th and > 90th percentiles were associated with significant risks of all-cause and cardiovascular mortality. In statin users, but not fibrate users, lower and higher levels of mean LDL-C and LDL-C-SD were both associated with elevated risks of all-cause and cardiovascular mortality. Conclusions The optimal level of LDL-C was found to be >90-103.59 mg/dL in T2D patients. Lower and higher levels of mean LDL-C and LDL-C-SD were associated with all-cause and cardiovascular mortality, revealing U-shaped associations. Further studies are necessary to validate the relationship between optimal LDL-C levels and all-cause and cardiovascular mortality in patients with diabetes.
Collapse
Affiliation(s)
- Chin-Huan Chang
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Shu-Tin Yeh
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Seng-Wei Ooi
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chung-Yi Li
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan City, Taiwan,Department of Public Health, College of Public Health, China Medical University, Taichung City, Taiwan,Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung City, Taiwan
| | - Hua-Fen Chen
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City, Taiwan,School of Medicine and Department of Public Health, College of Medicine, Fujen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
17
|
Abstract
Although human sperm is morphologically mature in the epididymis, it cannot fertilize eggs before capacitation. Cholesterol efflux from the sperm plasma membrane is a key molecular event essential for cytoplasmic alkalinization and hyperactivation, but the underlying mechanism remains unclear. The human voltage-gated proton (hHv1) channel functions as an acid extruder to regulate intracellular pHs of many cell types, including sperm. Aside from voltage and pH, Hv channels are also regulated by distinct ligands, such as Zn2+ and albumin. In the present work, we identified cholesterol as an inhibitory ligand of the hHv1 channel and further investigated the underlying mechanism using the single-molecule fluorescence resonance energy transfer (smFRET) approach. Our results indicated that cholesterol inhibits the hHv1 channel by stabilizing the voltage-sensing S4 segment at resting conformations, a similar mechanism also utilized by Zn2+. Our results suggested that the S4 segment is the central gating machinery in the hHv1 channel, on which voltage and distinct ligands are converged to regulate channel function. Identification of membrane cholesterol as an inhibitory ligand provides a mechanism by which the hHv1 channel regulates fertilization by linking the cholesterol efflux with cytoplasmic alkalinization, a change that triggers calcium influx through the CatSper channel. These events finally lead to hyperactivation, a remarkable change in the mobility pattern indicating fertilization competence of human sperm.
Collapse
|
18
|
Lazniewska J, Bader C, Hickey SM, Selemidis S, O'Leary J, Simpson PV, Stagni S, Plush SE, Massi M, Brooks D. Rhenium(I) conjugates as tools for tracking cholesterol in cells. Metallomics 2022; 14:mfac040. [PMID: 35657681 PMCID: PMC9344854 DOI: 10.1093/mtomcs/mfac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 11/15/2022]
Abstract
Cholesterol is vital to control membrane integrity and fluidity, but is also a precursor to produce steroid hormones, bile acids, and vitamin D. Consequently, altered cholesterol biology has been linked to many diseases, including metabolic syndromes and cancer. Defining the intracellular pools of cholesterol and its trafficking within cells is essential to understand both normal cell physiology and mechanisms of pathogenesis. We have synthesized a new cholesterol mimic (ReTEGCholestanol), comprising a luminescent rhenium metal complex and a cholestanol targeting unit, linked using a tetraethylene glycol (TEG) spacer. ReTEGCholestanol demonstrated favourable imaging properties and improved water solubility when compared to a cholesterol derivative, and structurally related probes lacking the TEG linker. A non-malignant and three malignant prostate cell lines were used to characterize the uptake and intracellular distribution of ReTEGCholestanol. The ReTEGCholestanol complex was effectively internalized and mainly localized to late endosomes/lysosomes in non-malignant PNT1a cells, while in prostate cancer cells it also accumulated in early endosomes and multivesicular bodies, suggesting disturbed cholesterol biology in the malignant cells. The ReTEGCholestanol is a novel imaging agent for visualizing endosomal uptake and trafficking, which may be used to define cholesterol related biology including membrane integration and altered lipid trafficking/processing.
Collapse
Affiliation(s)
- Joanna Lazniewska
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Christie Bader
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Shane M Hickey
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Stavros Selemidis
- Department of Human Biosciences, RMIT University, Melbourne, Victoria 3000, Australia
| | - John O'Leary
- Discipline of Histopathology, University of Dublin Trinity College, Dublin 2, Ireland
| | - Peter V Simpson
- School of Molecular and Life Sciences - Curtin University, Bentley, Western Australia 6102, Australia
| | - Stefano Stagni
- Department of Industrial Chemistry Toso Montanari, University of Bologna, Via Zamboni, 33, Bologna I-40136, Italy
| | - Sally E Plush
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Massimiliano Massi
- School of Molecular and Life Sciences - Curtin University, Bentley, Western Australia 6102, Australia
| | - Doug Brooks
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
- Discipline of Histopathology, University of Dublin Trinity College, Dublin 2, Ireland
- School of Molecular and Life Sciences - Curtin University, Bentley, Western Australia 6102, Australia
| |
Collapse
|
19
|
Wu Z, Jankowski V, Jankowski J. Irreversible post-translational modifications - Emerging cardiovascular risk factors. Mol Aspects Med 2022; 86:101010. [PMID: 34404548 DOI: 10.1016/j.mam.2021.101010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/19/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022]
Abstract
Despite the introduction of lipid-lowering drugs, antihypertensives, antiplatelet and anticoagulation therapies for primary prevention of cardiovascular and heart diseases (CVD), it remains the number one cause of death globally, raising the question for novel/further essential factors besides traditional risk factors such as cholesterol, blood pressure and coagulation. With continuous identification and characterization of non-enzymatic post-translationally modified isoforms of proteins and lipoproteins, it is becoming increasingly clear that irreversible non-enzymatic post-translational modifications (nPTMs) alter the biological functions of native proteins and lipoproteins thereby transforming innate serum components into CVD mediators. In particular renal insufficiency and metabolic imbalance are major contributors to the systemically increased concentration of reactive metabolites and thus increased frequency of nPTMs, promoting multi-morbid disease development centering around cardiovascular disease. nPTMs are significantly involved in the onset and progression of cardiovascular disease and represent a significant and novel risk factor. These insights represent potentially new avenues for risk assessment, prevention and therapy. This review chapter summarizes all forms of nPTMs found in CKD and under metabolic imbalance and discusses the biochemical connections between molecular alterations and the pathological impact on increased cardiovascular risk, novel nPTM-associated non-traditional cardiovascular risk factors, and clinical implication of nPTM in cardiovascular disease.
Collapse
Affiliation(s)
- Zhuojun Wu
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, the Netherlands.
| |
Collapse
|
20
|
Yao YF, Chen ZY, Luo TY, Dou XY, Chen HB. Cholesterol affects the relationship between albumin and major adverse cardiac events in patients with coronary artery disease: a secondary analysis. Sci Rep 2022; 12:12634. [PMID: 35879423 PMCID: PMC9314340 DOI: 10.1038/s41598-022-16963-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
We aimed to examine whether the efficacy of the risk of poor prognosis in patients with coronary artery disease is jointly affected by total cholesterol and baseline serum albumin in a secondary analysis of previous study. We analyzed the data of 204 patients from October 2014 to October 2017 for newly diagnosed stable CAD. The outcome was major adverse cardiac events (MACE; defined as all cause mortality, non fatal myocardial infarction, and non fatal stroke). The median duration of follow-up was 783 days. Multivariable COX model was performed to revalidate the relationship between the sALB and MACE and interaction tests were conducted to find the effects of total cholesterol on their association. A total of 28 MACE occurred among the 204 participants. The risk of MACE varied by baseline serum albumin and total cholesterol. Specifically, lower serum albumin indicated higher risk of MACE (HR 3.52, 95% CI 1.30-9.54), and a test for interaction between baseline serum albumin and total cholesterol on MACE was significant (P = 0.0005). We suggested that baseline serum albumin and total cholesterol could interactively affect the risk of poor prognosis of patients with coronary artery diseases. Our findings need to be confirmed by further randomized trials.
Collapse
Affiliation(s)
- Yu-Feng Yao
- Department of Ophthalmology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Shantou University Medical College, No. 22 Xinling Road, 15724172356, Shantou, 515031, Guangdong Province, China
| | - Zhen-Yu Chen
- Department of Cardiovascular Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Shantou University Medical College, No. 22 Xinling Road, 15724172356, Shantou, 515031, Guangdong Province, China
| | - Tian-Yi Luo
- Department of Ophthalmology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Shenzhen University, Shenzhen, 518037, Guangdong Province, China
| | - Xiao-Yan Dou
- Department of Ophthalmology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China.
| | - Hai-Bo Chen
- Department of Cardiovascular Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China.
| |
Collapse
|
21
|
Schekatolina S, Lahovska V, Bekshaev A, Kontush S, Le Goff W, Kontush A. Mathematical Modelling of Material Transfer to High-Density Lipoprotein (HDL) upon Triglyceride Lipolysis by Lipoprotein Lipase: Relevance to Cardioprotective Role of HDL. Metabolites 2022; 12:metabo12070623. [PMID: 35888747 PMCID: PMC9317498 DOI: 10.3390/metabo12070623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/14/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
High-density lipoprotein (HDL) contributes to lipolysis of triglyceride-rich lipoprotein (TGRL) by lipoprotein lipase (LPL) via acquirement of surface lipids, including free cholesterol (FC), released upon lipolysis. According to the reverse remnant-cholesterol transport (RRT) hypothesis recently developed by us, acquirement of FC by HDL is reduced at both low and extremely high HDL concentrations, potentially underlying the U-shaped relationship between HDL-cholesterol and cardiovascular disease. Mechanisms underlying impaired FC transfer however remain indeterminate. We developed a mathematical model of material transfer to HDL upon TGRL lipolysis by LPL. Consistent with experimental observations, mathematical modelling showed that surface components of TGRL, including FC, were accumulated in HDL upon lipolysis. The modelling successfully reproduced major features of cholesterol accumulation in HDL observed experimentally, notably saturation of this process over time and appearance of a maximum as a function of HDL concentration. The calculations suggested that the both phenomena resulted from competitive fluxes of FC through the HDL pool, including primarily those driven by FC concentration gradient between TGRL and HDL on the one hand and mediated by lecithin-cholesterol acyltransferase (LCAT) and cholesteryl ester transfer protein (CETP) on the other hand. These findings provide novel opportunities to revisit our view of HDL in the framework of RRT.
Collapse
Affiliation(s)
| | - Viktoriia Lahovska
- Odessa National Technological University, 65000 Odessa, Ukraine; (S.S.); (V.L.)
| | - Aleksandr Bekshaev
- Physics Research Institute, I.I. Mechnikov Odessa National University, 65082 Odessa, Ukraine; (A.B.); (S.K.)
| | - Sergey Kontush
- Physics Research Institute, I.I. Mechnikov Odessa National University, 65082 Odessa, Ukraine; (A.B.); (S.K.)
| | - Wilfried Le Goff
- Unité de Recherche sur les Maladies Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale (INSERM), le Métabolisme et la Nutrition, ICAN, Sorbonne Université, F-75013 Paris, France;
| | - Anatol Kontush
- Unité de Recherche sur les Maladies Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale (INSERM), le Métabolisme et la Nutrition, ICAN, Sorbonne Université, F-75013 Paris, France;
- Correspondence: ; Tel.: +33-(1)-40-77-96-33; Fax: +33-(1)-40-77-96-45
| |
Collapse
|
22
|
Ward ES, Gelinas D, Dreesen E, Van Santbergen J, Andersen JT, Silvestri NJ, Kiss JE, Sleep D, Rader DJ, Kastelein JJP, Louagie E, Vidarsson G, Spriet I. Clinical Significance of Serum Albumin and Implications of FcRn Inhibitor Treatment in IgG-Mediated Autoimmune Disorders. Front Immunol 2022; 13:892534. [PMID: 35757719 PMCID: PMC9231186 DOI: 10.3389/fimmu.2022.892534] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/22/2022] [Indexed: 12/26/2022] Open
Abstract
Serum albumin (SA), the most abundant soluble protein in the body, maintains plasma oncotic pressure and regulates the distribution of vascular fluid and has a range of other important functions. The goals of this review are to expand clinical knowledge regarding the functions of SA, elucidate effects of dysregulated SA concentration, and discuss the clinical relevance of hypoalbuminemia resulting from various diseases. We discuss potential repercussions of SA dysregulation on cholesterol levels, liver function, and other processes that rely on its homeostasis, as decreased SA concentration has been shown to be associated with increased risk for cardiovascular disease, hyperlipidemia, and mortality. We describe the anti-inflammatory and antioxidant properties of SA, as well as its ability to bind and transport a plethora of endogenous and exogenous molecules. SA is the primary serum protein involved in binding and transport of drugs and as such has the potential to affect, or be affected by, certain medications. Of current relevance are antibody-based inhibitors of the neonatal Fc receptor (FcRn), several of which are under clinical development to treat immunoglobulin G (IgG)-mediated autoimmune disorders; some have been shown to decrease SA concentration. FcRn acts as a homeostatic regulator of SA by rescuing it, as well as IgG, from intracellular degradation via a common cellular recycling mechanism. Greater clinical understanding of the multifunctional nature of SA and the potential clinical impact of decreased SA are needed; in particular, the potential for certain treatments to reduce SA concentration, which may affect efficacy and toxicity of medications and disease progression.
Collapse
Affiliation(s)
- E Sally Ward
- Cancer Sciences Unit, Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | | | - Erwin Dreesen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | | | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Pharmacology, University of Oslo, Oslo, Norway
| | | | - Joseph E Kiss
- Vitalant Northeast Division and Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Daniel J Rader
- Departments of Genetics and Medicine, Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John J P Kastelein
- Department of Vascular Medicine, Genetics of Cardiovascular Disease, Academic Medical Center (AMC) of the University of Amsterdam, Amsterdam, Netherlands
| | | | - Gestur Vidarsson
- Department of Experimental Immunohematology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Isabel Spriet
- Department of Clinical Pharmacology and Pharmacotherapy, KU Leuven, Leuven, Belgium.,Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Different Pathways of Cellular Cholesterol Efflux. Cell Biochem Biophys 2022; 80:471-481. [PMID: 35737216 DOI: 10.1007/s12013-022-01081-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 12/22/2022]
Abstract
Cholesterol efflux is the first and rate-limiting step of reverse cholesterol transport (RCT) from peripheric cells to the liver. The involvement of high-density lipoprotein (HDL) in RCT determines the atheroprotective properties of HDL. Cholesterol efflux from different membrane pools includes both passive and energy-dependent processes. The first type of route consists of cholesterol desorption from the cell membrane into the unstirred layer adjacent to the cell surface and diffusion in the water phase. Moreover, the selective uptake and facilitated diffusion of cholesterol and cholesteryl ester molecules through the hydrophobic tunnel in the scavenger receptor BI molecule does not require energy consumption. The second type of route includes active cholesterol export by the ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1). Several cholesterol acceptors specifically bind cholesterol and phospholipid molecules, and cholesterol binding to the albumin molecule, which acts as a shuttle, significantly increases cholesterol movement between acceptors and red blood cells, thus functioning as a sink for cholesterol. Cholesterol and phospholipid molecules effluxed from macrophages by ABCA1 are accepted exclusively by the lipid-free apolipoprotein apoA-I, which is the major protein moiety of HDL, whereas those effluxed by ABCG1 are accepted by HDL. ABCA1- and ABCG1-mediated cholesterol transport, together with cholesterol diffusion, largely determine cholesterol turnover at the physiological level of intracellular cholesterol. However, at cholesterol overload, ABCA1-mediated efflux prevails over other routes. The exchange of apoA-I between lipid-free and lipid-associated states and the synergism of nascent and mature HDL contribute to cholesterol efflux efficiency. Moreover, extracellular cholesterol deposits and microvesicles may be involved in RCT.
Collapse
|
24
|
He J, Zhou X, Xu F, He H, Ma S, Liu X, Zhang M, Zhang W, Liu J. Anchoring β-CD on simvastatin-loaded rHDL for selective cholesterol crystals dissolution and enhanced anti-inflammatory effects in macrophage/foam cells. Eur J Pharm Biopharm 2022; 174:144-154. [PMID: 35447349 DOI: 10.1016/j.ejpb.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/01/2022] [Accepted: 04/14/2022] [Indexed: 12/23/2022]
Abstract
Macrophage/foam cells and cholesterol crystals (CCs) have been regarded as the central triggers of maladaptive inflammation in atherosclerotic plaque. Despite the tremendous progress of recombinant high-density lipoprotein (rHDL) serving for targeted drug delivery to alleviate inflammation in macrophage/foam cells, the active attempt to modulate/improve its CCs dissolution capacity remains poorly explored. The untreated CCs can seriously aggravate inflammation and threaten plaque stability. Based on the superb ability of β-cyclodextrin (β-CD) to bind CCs and promote cholesterol efflux, simvastatin-loaded discoidal-rHDL (ST-d-rHDL) anchored with β-CD (βCD-ST-d-rHDL) was constructed. We verified that βCD-ST-d-rHDL specifically bound and dissolved CCs extracellularly and intracellularly. Furthermore, anchoring β-CD onto the surface of ST-d-rHDL enhanced its cholesterol removal ability in RAW 264.7 cell-derived foam cells characterized by accelerated cholesterol efflux, reduced intracellular lipid deposition, and improved cell membrane fluidity/permeability. Finally, βCD-ST-d-rHDL exerted efficient drug delivery and effective anti-inflammatory effects in macrophage/foam cells. Collectively, anchoring β-CD onto the surface of ST-d-rHDL for selective CCs dissolution, accelerated cholesterol efflux, and improved drug delivery represents an effective strategy to enhance anti-inflammatory effects for the therapy of atherosclerosis.
Collapse
Affiliation(s)
- Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Xiaoju Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China; Institute of Pharmaceutics, Nanjing Research Center, Jiangsu Chia-tai Tianqing Pharmaceutical Co. , Ltd., Nanjing, Jiangsu 210008, PR China
| | - Fengfei Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Hongliang He
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Shuangyan Ma
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Xinyue Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Mengyuan Zhang
- Department of Pharmaceutical Engineering, Jiangsu Food & Pharmaceutical Science College, Huaian, Jiangsu 223003, PR China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China.
| |
Collapse
|
25
|
Sterolight as imaging tool to study sterol uptake, trafficking and efflux in living cells. Sci Rep 2022; 12:6264. [PMID: 35428843 PMCID: PMC9012876 DOI: 10.1038/s41598-022-10134-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/25/2022] [Indexed: 12/19/2022] Open
Abstract
Information about cholesterol subcellular localization and transport pathways inside cells is essential for understanding and treatment of cholesterol-related diseases. However, there is a lack of reliable tools to monitor it. This work follows the fate of Sterolight, a BODIPY-labelled sterol, within the cell and demonstrates it as a suitable probe for visualization of sterol/lipid trafficking. Sterolight enters cells through an energy-independent process and knockdown experiments suggest caveolin-1 as its potential cellular carrier. Intracellular transport of Sterolight is a rapid process, and transfer from ER and mitochondria to lysosomes and later to lipid droplets requires the participation of active microtubules, as it can be inhibited by the microtubule disruptor nocodazole. Excess of the probe is actively exported from cells, in addition to being stored in lipid droplets, to re-establish the sterol balance. Efflux occurs through a mechanism requiring energy and may be selectively poisoned with verapamil or blocked in cells with mutated cholesterol transporter NPC1. Sterolight is efficiently transferred within and between different cell populations, making it suitable for monitoring numerous aspects of sterol biology, including the live tracking and visualization of intracellular and intercellular transport.
Collapse
|
26
|
Juhl AD, Wüstner D. Pathways and Mechanisms of Cellular Cholesterol Efflux-Insight From Imaging. Front Cell Dev Biol 2022; 10:834408. [PMID: 35300409 PMCID: PMC8920967 DOI: 10.3389/fcell.2022.834408] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
Cholesterol is an essential molecule in cellular membranes, but too much cholesterol can be toxic. Therefore, mammalian cells have developed complex mechanisms to remove excess cholesterol. In this review article, we discuss what is known about such efflux pathways including a discussion of reverse cholesterol transport and formation of high-density lipoprotein, the function of ABC transporters and other sterol efflux proteins, and we highlight their role in human diseases. Attention is paid to the biophysical principles governing efflux of sterols from cells. We also discuss recent evidence for cholesterol efflux by the release of exosomes, microvesicles, and migrasomes. The role of the endo-lysosomal network, lipophagy, and selected lysosomal transporters, such as Niemann Pick type C proteins in cholesterol export from cells is elucidated. Since oxysterols are important regulators of cellular cholesterol efflux, their formation, trafficking, and secretion are described briefly. In addition to discussing results obtained with traditional biochemical methods, focus is on studies that use established and novel bioimaging approaches to obtain insight into cholesterol efflux pathways, including fluorescence and electron microscopy, atomic force microscopy, X-ray tomography as well as mass spectrometry imaging.
Collapse
Affiliation(s)
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
27
|
Römer A, Rawat D, Linn T, Petry SF. Preparation of fatty acid solutions exerts significant impact on experimental outcomes in cell culture models of lipotoxicity. Biol Methods Protoc 2022; 7:bpab023. [PMID: 35036572 PMCID: PMC8754478 DOI: 10.1093/biomethods/bpab023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/14/2022] Open
Abstract
Free fatty acids are essentially involved in the pathogenesis of chronic diseases such as diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular disease. They promote mitochondrial dysfunction, oxidative stress, respiratory chain uncoupling, and endoplasmic reticulum stress and modulate stress-sensitive pathways. These detrimental biological effects summarized as lipotoxicity mainly depend on fatty acid carbon chain length, degree of unsaturation, concentration, and treatment time. Preparation of fatty acid solutions involves dissolving and complexing. Solvent toxicity and concentration, amount of bovine serum albumin (BSA), and ratio of albumin to fatty acids can vary significantly between equal concentrations, mediating considerable harmful effects and/or interference with certain assays such as 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). Herein, we studied the impact of commonly used solvents ethanol and dimethyl sulfoxide and varying concentrations of BSA directly and in solution with oleic acid on MTT to formazan conversion, adenosine triphosphate level, and insulin content and secretion of murine β-cell line MIN6 employing different treatment duration. Our data show that experimental outcomes and assay readouts can be significantly affected by mere preparation of fatty acid solutions and should thus be carefully considered and described in detail to ensure comparability and distinct evaluation of data.
Collapse
Affiliation(s)
- Axel Römer
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Divya Rawat
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Sebastian F Petry
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
28
|
Li D, Ruan Z, Wu B. Association of Red Blood Cell Distribution Width-Albumin Ratio for Acute Myocardial Infarction Patients with Mortality: A Retrospective Cohort Study. Clin Appl Thromb Hemost 2022; 28:10760296221121286. [PMID: 36045634 PMCID: PMC9445528 DOI: 10.1177/10760296221121286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Red blood cell distribution width (RDW) was a risk factor for poor prognosis in acute myocardial infarction (AMI). Recent reports suggested that combining RDW with other laboratory metrics could provide a better prediction. This retrospective study aimed to investigate whether the RDW-albumin ratio (RAR) may be associated with mortality after an AMI. METHODS This cohort study was conducted among adults (over 16 years old) with AMI in the Medical Information Mart for Intensive Care Database III V1.4 (MIMIC-III). The primary outcome was 30-day mortality, and the secondary outcome was 1-year and 3-year mortality. Cox hazard regression model and Kaplan-Meier survival curves were constructed to estimate the effect of biomarkers on mortality. We used three models to adjust for potential bias. Receiver operating characteristic (ROC) curves and the area under the curve (AUC) were analyzed for the excellent performance of RAR on prognosis. RESULTS A total of 826 patients were eventually enrolled in our study. In multivariate analysis, RAR was found to be associated with 30-day mortality (Model 3: HR = 1.23, 95% CI = 1.09-1.39, P < .001). In addition, Subgroup analysis showed that the effect of RAR was higher in female patients than in male patients (P for interaction = .026). Kaplan-Meier survival curves showed that patients in the lower RAR quartile tended to have higher survival rates in the short and long term. AMI patients with RAR ≥ 4 had a 122% increase in 3-year mortality. Results of ROC and AUC showed that the prognostic performance of RAR for mortality was the best (30-day mortality: 0.703; 1-year mortality: 0.729; 3-year mortality: 0.737). CONCLUSIONS RAR is a simple and stable predictor of prognosis in AMI patients. Our results support RAR = 4.0 as a criterion for prognostic risk stratification of AMI patients.
Collapse
Affiliation(s)
- Dan Li
- The First Clinical College, Shandong Chinese Medical University, Ji Nan, People’s Republic of China
| | - Zhishen Ruan
- The First Clinical College, Shandong Chinese Medical University, Ji Nan, People’s Republic of China
| | - Bo Wu
- The First Clinical College, Shandong Chinese Medical University, Ji Nan, People’s Republic of China
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
- Bo Wu, Department of Cardiovascular Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 42 Wenhua West Road, Ji Nan city, Shandong Province, People’s Republic of China.
| |
Collapse
|
29
|
Zheng A, Dubuis G, Georgieva M, Ferreira CSM, Serulla M, Del Carmen Conde Rubio M, Trofimenko E, Mercier T, Decosterd L, Widmann C. HDLs extract lipophilic drugs from cells. J Cell Sci 2021; 135:273878. [PMID: 34981808 PMCID: PMC8919334 DOI: 10.1242/jcs.258644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022] Open
Abstract
High-density lipoproteins (HDLs) prevent cell death induced by a variety of cytotoxic drugs. The underlying mechanisms are however still poorly understood. Here, we present evidence that HDLs efficiently protect cells against thapsigargin (TG), a sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) inhibitor, by extracting the drug from cells. Drug efflux could also be triggered to some extent by low-density lipoproteins and serum. HDLs did not reverse the non-lethal mild ER stress response induced by low TG concentrations or by SERCA knockdown, but HDLs inhibited the toxic SERCA-independent effects mediated by high TG concentrations. HDLs could extract other lipophilic compounds, but not hydrophilic substances. This work shows that HDLs utilize their capacity of loading themselves with lipophilic compounds, akin to their ability to extract cellular cholesterol, to reduce the cell content of hydrophobic drugs. This can be beneficial if lipophilic xenobiotics are toxic but may be detrimental to the therapeutic benefit of lipophilic drugs such as glibenclamide. Summary: HDLs, akin to their capacity for extracting cholesterol, can remove lipophilic compounds from cells, thus protecting the cells when these compounds are toxic.
Collapse
Affiliation(s)
- Adi Zheng
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Gilles Dubuis
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Maria Georgieva
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | | | - Marc Serulla
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | | | - Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Thomas Mercier
- Laboratory of Clinical Pharmacology, Lausanne University Hospital (CHUV) and University of Lausanne, Switzerland
| | - Laurent Decosterd
- Laboratory of Clinical Pharmacology, Lausanne University Hospital (CHUV) and University of Lausanne, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| |
Collapse
|
30
|
Lien KA, Dinshaw K, Nichols RJ, Cassidy-Amstutz C, Knight M, Singh R, Eltis LD, Savage DF, Stanley SA. A nanocompartment system contributes to defense against oxidative stress in Mycobacterium tuberculosis. eLife 2021; 10:e74358. [PMID: 34751132 PMCID: PMC8635971 DOI: 10.7554/elife.74358] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/06/2021] [Indexed: 01/18/2023] Open
Abstract
Encapsulin nanocompartments are an emerging class of prokaryotic protein-based organelle consisting of an encapsulin protein shell that encloses a protein cargo. Genes encoding nanocompartments are widespread in bacteria and archaea, and recent works have characterized the biochemical function of several cargo enzymes. However, the importance of these organelles to host physiology is poorly understood. Here, we report that the human pathogen Mycobacterium tuberculosis (Mtb) produces a nanocompartment that contains the dye-decolorizing peroxidase DyP. We show that this nanocompartment is important for the ability of Mtb to resist oxidative stress in low pH environments, including during infection of host cells and upon treatment with a clinically relevant antibiotic. Our findings are the first to implicate a nanocompartment in bacterial pathogenesis and reveal a new mechanism that Mtb uses to combat oxidative stress.
Collapse
Affiliation(s)
- Katie A Lien
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, BerkeleyBerkeleyUnited States
| | - Kayla Dinshaw
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, BerkeleyBerkeleyUnited States
| | - Robert J Nichols
- Department of Molecular and Cell Biology, Division of Biochemistry, Biophysics and Structural Biology, University of California, BerkeleyBerkeleyUnited States
| | - Caleb Cassidy-Amstutz
- Department of Molecular and Cell Biology, Division of Biochemistry, Biophysics and Structural Biology, University of California, BerkeleyBerkeleyUnited States
| | - Matthew Knight
- Department of Plant and Microbial Biology, University of California, BerkeleyBerkeleyUnited States
| | - Rahul Singh
- Department of Microbiology and Immunology, The University of British ColumbiaVancouverCanada
| | - Lindsay D Eltis
- Department of Microbiology and Immunology, The University of British ColumbiaVancouverCanada
| | - David F Savage
- Department of Molecular and Cell Biology, Division of Biochemistry, Biophysics and Structural Biology, University of California, BerkeleyBerkeleyUnited States
| | - Sarah A Stanley
- Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, BerkeleyBerkeleyUnited States
- School of Public Health, Division of Infectious Diseases and Vaccinology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
31
|
Darabi M, Kontush A. High-density lipoproteins (HDL): Novel function and therapeutic applications. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159058. [PMID: 34624514 DOI: 10.1016/j.bbalip.2021.159058] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 12/30/2022]
Abstract
The failure of high-density lipoprotein (HDL)-raising agents to reduce cardiovascular disease (CVD) together with recent findings of increased cardiovascular mortality in subjects with extremely high HDL-cholesterol levels provide new opportunities to revisit our view of HDL. The concept of HDL function developed to explain these contradictory findings has recently been expanded by a role played by HDL in the lipolysis of triglyceride-rich lipoproteins (TGRLs) by lipoprotein lipase. According to the reverse remnant-cholesterol transport (RRT) hypothesis, HDL critically contributes to TGRL lipolysis via acquirement of surface lipids, including free cholesterol, released from TGRL. Ensuing cholesterol transport to the liver with excretion into the bile may reduce cholesterol influx in the arterial wall by accelerating removal from circulation of atherogenic, cholesterol-rich TGRL remnants. Such novel function of HDL opens wide therapeutic applications to reduce CVD in statin-treated patients, which primarily involve activation of cholesterol flux upon lipolysis.
Collapse
Affiliation(s)
- Maryam Darabi
- National Institute for Health and Medical Research (INSERM), UMRS 1166 ICAN, Faculty of Medicine Pitié-Salpêtrière, Sorbonne University, Paris, France
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMRS 1166 ICAN, Faculty of Medicine Pitié-Salpêtrière, Sorbonne University, Paris, France.
| |
Collapse
|
32
|
de Araújo Lira AL, de Fátima Mello Santana M, de Souza Pinto R, Minanni CA, Iborra RT, de Lima AMS, Correa-Giannella ML, Passarelli M, Queiroz MS. Serum albumin modified by carbamoylation impairs macrophage cholesterol efflux in diabetic kidney disease. J Diabetes Complications 2021; 35:107969. [PMID: 34183248 DOI: 10.1016/j.jdiacomp.2021.107969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/05/2021] [Accepted: 05/23/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Abnormalities in lipid metabolism, accumulation of uremic toxins and advanced glycation end products may contribute to worsening atherosclerosis. This study characterized the glycation and carbamoylation profile of serum albumin isolated from individuals with diabetic kidney disease and its influence on cholesterol efflux. MATERIAL AND METHODS 49 patients with type 2 diabetes (T2DM) and different eGFR evaluated glycation and carbamoylation profile by measurement of carboxymethyl lysine (CML) and carbamoylated proteins (CBL) in plasma by ELISA, homocitrulline (HCit) in plasma by colorimetry. In the isolated albumins, we quantified CBL (ELISA) and total AGE and pentosidine by fluorescence. Macrophages were treated with albumin isolated, and 14C-Cholesterol efflux mediated by HDL2 or HDL3 was measured. Kruskal-Wallis test, Jonckheere-Terpstra test and Brunner's posttest were used for comparisons among groups. RESULTS Determination of CML, HCit, CBL in plasma, as total AGE and pentosidine in albumins, did not differ between groups; however, CBL in the isolated albumins was higher in the more advanced stages of CKD (p=0.0414). There was reduction in the 14C-cholesterol efflux after treatment for 18h with albumin isolated from patients with eGFR<60mL/min/1.73m2 compared with control group mediated by HDL2 (p=0.0288) and HDL3 (p<0.0001), as well as when compared with eGFR ≥60mL/min/1.73m2 per HDL2 (p=0.0001) and HDL3 (p<0.0001). Treatment for 48h showed that eGFR<15mL/min/1.73m2 had a lower percentage of 14C-cholesterol efflux mediated by HDL2 compared to control and other CKD groups (p=0.0274). CONCLUSIONS Albumins isolated from individuals with T2DM and eGFR<60mL/min/1.73m2 suffer greater carbamoylation, and they impair the cholesterol efflux mediated by HDL2 and HDL3. In turn, this could promote lipids accumulation in macrophages and disorders in reverse cholesterol transport.
Collapse
Affiliation(s)
| | | | - Raphael de Souza Pinto
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil
| | - Carlos André Minanni
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil
| | - Rodrigo Tallada Iborra
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil; Sao Judas Tadeu University, Sao Paulo, Brazil
| | - Adriana Machado Saldiba de Lima
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil; Sao Judas Tadeu University, Sao Paulo, Brazil
| | - Maria Lúcia Correa-Giannella
- Laboratory of Carbohydrates and Radioimuneassays (LIM 18), Clinical Hospital, Medical School, University of Sao Paulo, Sao Paulo, Brazil; Department of Graduation in Medicine, Nove de Julho University (Uninove), Sao Paulo, Brazil
| | - Marisa Passarelli
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil; Department of Graduation in Medicine, Nove de Julho University (Uninove), Sao Paulo, Brazil
| | - Márcia Silva Queiroz
- Endocrinology Division, Internal Medicine Department, University of Sao Paulo Medical School, Sao Paulo, Brazil; Department of Graduation in Medicine, Nove de Julho University (Uninove), Sao Paulo, Brazil.
| |
Collapse
|
33
|
Escolà-Gil JC, Rotllan N, Julve J, Blanco-Vaca F. Reverse Cholesterol Transport Dysfunction Is a Feature of Familial Hypercholesterolemia. Curr Atheroscler Rep 2021; 23:29. [PMID: 33914189 DOI: 10.1007/s11883-021-00928-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 02/21/2023]
Abstract
PURPOSE OF REVIEW We seek to establish whether high-density lipoprotein HDL metabolism and reverse cholesterol transport (RCT) impairment is an intrinsic feature of familial hypercholesterolemia (FH). RECENT FINDINGS RCT from macrophages (m-RCT), a vascular cell type of major influence on atherosclerosis, is impaired in FH due to defective low-density lipoprotein receptor (LDLR) function via both the HDL- and LDL-mediated pathways. Potential mechanisms include impaired HDL metabolism, which is linked to increased LDL levels, as well as the increased transport of cellular unesterified cholesterol to LDL, which presents a defective catabolism. RCT dysfunction is consistently associated with mutation-positive FH linked to decreased HDL levels as well as impaired HDL remodeling and LDLR function. It remains to be explored whether these alterations are also present in less well-characterized forms of FH, such as cases with no identified mutations, and whether they are fully corrected by current standard treatments.
Collapse
Affiliation(s)
- Joan Carles Escolà-Gil
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain. .,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain. .,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Noemí Rotllan
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Josep Julve
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco Blanco-Vaca
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain
| |
Collapse
|
34
|
He J, Yang Y, Zhou X, Zhang W, Liu J. Shuttle/sink model composed of β-cyclodextrin and simvastatin-loaded discoidal reconstituted high-density lipoprotein for enhanced cholesterol efflux and drug uptake in macrophage/foam cells. J Mater Chem B 2021; 8:1496-1506. [PMID: 31999290 DOI: 10.1039/c9tb02101a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Targeting drug delivery to macrophage/foam cells is challenged owing to the poor cell permeability and fluidity resulting from the massive accumulation of intracellular cholesterol in atherosclerosis (AS). Discoidal reconstituted high-density lipoprotein (d-rHDL) has been well regarded as a potential drug delivery system for AS by virtue of its plaque-targeting and cholesterol removal abilities, while the latter is compromised by the high activation energy of cholesterol efflux. It is reported that a low concentration of β-cyclodextrin (β-CD) can function as a cholesterol shuttle to promote cholesterol efflux from cells to the extracellular acceptors (cholesterol sink, such as HDL particles), but it is still unknown whether the combination of β-CD with a drug-loaded d-rHDL can function as a shuttle/sink model to promote the remodeling and drug release of the d-rHDL carrier after accelerating the cholesterol efflux. Furthermore, it is interesting to investigate whether enhanced cholesterol efflux can improve the cellular drug uptake by restoring the permeability and fluidity of the cell membrane. Here, simvastatin-loaded d-rHDL (ST-d-rHDL) was combined with different concentrations of β-CD. Compared with ST-d-rHDL alone, the cholesterol removal ability of ST-d-rHDL combined with 0.5 mM of β-CD increased by 31-fold after incubation for 6 h and the cumulative drug release of ST-d-rHDL increased by two-fold during the initial 1 h in an acellular mimetic system. In macrophage/foam cells, 0.5 mM of β-CD showed superior promoting effects in the cholesterol removal ability and remodeling of ST-d-rHDL compared to 0.1 mM of β-CD. The high concentration of β-CD at 2 mM displayed a low efficiency for accelerating cholesterol efflux, which might function as a cholesterol sink rather than a cholesterol shuttle. Moreover, the permeability and fluidity of the cell membrane were improved by combining 0.5 mM of β-CD with ST-d-rHDL, which exhibited an enhanced cellular drug uptake and inhibiting effect on the intracellular lipid deposition and secretion of inflammatory cytokine. Collectively, combination of β-CD and ST-d-rHDL as a shuttle/sink model could enhance cholesterol efflux and drug uptake to suppress inflammation in macrophage/foam cells.
Collapse
Affiliation(s)
- Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Yun Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China. and State Key Laboratory of Long-acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co., Ltd, Yantai, Shandong 264670, P. R. China
| | - Xiaoju Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China. and Institute of Pharmaceutics, Nanjing Research Center, Jiangsu Chia-tai Tianqing Pharmaceutical Co., Ltd, Nanjing, Jiangsu 210008, P. R. China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| |
Collapse
|
35
|
Pedrelli M, Parini P, Kindberg J, Arnemo JM, Bjorkhem I, Aasa U, Westerståhl M, Walentinsson A, Pavanello C, Turri M, Calabresi L, Öörni K, Camejo G, Fröbert O, Hurt-Camejo E. Vasculoprotective properties of plasma lipoproteins from brown bears (Ursus arctos). J Lipid Res 2021; 62:100065. [PMID: 33713671 PMCID: PMC8131316 DOI: 10.1016/j.jlr.2021.100065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022] Open
Abstract
Plasma cholesterol and triglyceride (TG) levels are twice as high in hibernating brown bears (Ursus arctos) than healthy humans. Yet, bears display no signs of early stage atherosclerosis development when adult. To explore this apparent paradox, we analyzed plasma lipoproteins from the same 10 bears in winter (hibernation) and summer using size exclusion chromatography, ultracentrifugation, and electrophoresis. LDL binding to arterial proteoglycans (PGs) and plasma cholesterol efflux capacity (CEC) were also evaluated. The data collected and analyzed from bears were also compared with those from healthy humans. In bears, the cholesterol ester, unesterified cholesterol, TG, and phospholipid contents of VLDL and LDL were higher in winter than in summer. The percentage lipid composition of LDL differed between bears and humans but did not change seasonally in bears. Bear LDL was larger, richer in TGs, showed prebeta electrophoretic mobility, and had 5-10 times lower binding to arterial PGs than human LDL. Finally, plasma CEC was higher in bears than in humans, especially the HDL fraction when mediated by ABCA1. These results suggest that in brown bears the absence of early atherogenesis is likely associated with a lower affinity of LDL for arterial PGs and an elevated CEC of bear plasma.
Collapse
Affiliation(s)
- Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Paolo Parini
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Metabolism Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Infection, Karolinska university Hospital, Stockholm, Sweden
| | - Jonas Kindberg
- Norwegian Institute for Nature Research, Trondheim, Norway; Swedish University of Agricultural Sciences, Department of Wildlife, Fish, and Environmental Studies, Umeå, Sweden
| | - Jon M Arnemo
- Swedish University of Agricultural Sciences, Department of Wildlife, Fish, and Environmental Studies, Umeå, Sweden; Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, Campus Evenstad, Koppang, Norway
| | - Ingemar Bjorkhem
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Aasa
- Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden
| | - Maria Westerståhl
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Walentinsson
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Chiara Pavanello
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marta Turri
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Laura Calabresi
- Centro Enrica Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Gérman Camejo
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ole Fröbert
- Swedish University of Agricultural Sciences, Department of Wildlife, Fish, and Environmental Studies, Umeå, Sweden; Örebro University, Faculty of Health, Department of Cardiology, Örebro, Sweden
| | - Eva Hurt-Camejo
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
36
|
Takahara M, Mochizuki S, Wakabayashi R, Minamihata K, Goto M, Sakurai K, Kamiya N. Extending the Half-Life of a Protein in Vivo by Enzymatic Labeling with Amphiphilic Lipopeptides. Bioconjug Chem 2021; 32:655-660. [PMID: 33689283 DOI: 10.1021/acs.bioconjchem.1c00027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synthesis of lipid-protein conjugates is one of the significant techniques in drug delivery systems of proteins; however, the intact conjugation of a lipid and protein is yet challenging due to the hydrophobicity of lipid molecules. In order to facilitate easy handling of the lipid moiety in conjugation, we have focused on a microbial transglutaminase (MTG) that can ligate specific lysine (K) and glutamine (Q) residues in lipopeptides and a protein of interest. As MTG substrates, monolipid- and dilipid-fused amphiphilic short lipopeptide substrates (lipid-G3S-RHK or lipid2-KG3S-RHK) were designed. These amphiphilic lipopeptides and a model protein (enhanced green fluorescent protein, EGFP) fused with LLQG (LQ-EGFP) were both water-soluble, and thus lipid-protein conjugates were efficiently obtained through the MTG reaction with a >80% conversion rate of LQ-EGFP even using cholesterol-G3S-RHK. In vitro cell adhesion and in vivo half-life stability of the successfully obtained lipid-protein conjugates were evaluated, showing that the monocholesterol-G3S-RHK modification of a protein gave the highest cell adhesion efficiency and longest half-life time by formation of a stable albumin/lipid-protein complex.
Collapse
Affiliation(s)
- Mari Takahara
- Department of Materials Science & Chemical Engineering, National Institute of Technology, Kitakyushu College, 5-20-1 Shii, Kokuraminamiku, Kitakyushu 802-0985, Japan
| | - Shinichi Mochizuki
- Department of Chemistry and Biochemistry, the University of Kitakyushu, 1-1 Hibikino, Wakamatsuku, Kitakyushu 808-0135, Japan
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan.,Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, the University of Kitakyushu, 1-1 Hibikino, Wakamatsuku, Kitakyushu 808-0135, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan.,Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| |
Collapse
|
37
|
Harrison RE, Brown MR, Strand MR. Whole blood and blood components from vertebrates differentially affect egg formation in three species of anautogenous mosquitoes. Parasit Vectors 2021; 14:119. [PMID: 33627180 PMCID: PMC7905675 DOI: 10.1186/s13071-021-04594-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Background Most female mosquitoes are anautogenous and must blood feed on a vertebrate host to produce eggs. Prior studies show that the number of eggs females lay per clutch correlates with the volume of blood ingested and that protein is the most important macronutrient for egg formation. In contrast, how whole blood, blood fractions and specific blood proteins from different vertebrates affect egg formation is less clear. Since egg formation is best understood in Aedes aegypti, we examined how blood and blood components from different vertebrates affect this species and two others: the malaria vector Anopheles gambiae and arbovirus vector Culex quinquefasciatus. Methods Adult female mosquitoes were fed blood, blood fractions and purified major blood proteins from different vertebrate hosts. Markers of reproductive response including ovary ecdysteroidogenesis, yolk deposition into oocytes and number of mature eggs produced were measured. Results Ae. aegypti, An. gambiae and C. quinquefasciatus responded differently to meals of whole blood, plasma or blood cells from human, rat, chicken and turkey hosts. We observed more similarities between the anthropophiles Ae. aegypti and An. gambiae than the ornithophile C. quinquefasciatus. Focusing on Ae. aegypti, the major plasma-derived proteins (serum albumin, fibrinogen and globulins) differentially stimulated egg formation as a function of vertebrate host source. The major blood cell protein, hemoglobin, stimulated yolk deposition when from pigs but not humans, cows or sheep. Serum albumins from different vertebrates also variably affected egg formation. Bovine serum albumin (BSA) stimulated ovary ecdysteroidogenesis, but more weakly induced digestive enzyme activities than whole blood. In contrast, BSA-derived peptides and free amino acids had no stimulatory effects on ecdysteroidogenesis or yolk deposition into oocytes. Conclusions Whole blood, blood fractions and specific blood proteins supported egg formation in three species of anautogenous mosquitoes but specific responses varied with the vertebrate source of the blood components tested.![]()
Collapse
Affiliation(s)
- Ruby E Harrison
- Department of Entomology, The University of Georgia, 120 Cedar Street, 420 Biological Sciences, Athens, GA, 30602, USA
| | - Mark R Brown
- Department of Entomology, The University of Georgia, 120 Cedar Street, 420 Biological Sciences, Athens, GA, 30602, USA
| | - Michael R Strand
- Department of Entomology, The University of Georgia, 120 Cedar Street, 420 Biological Sciences, Athens, GA, 30602, USA.
| |
Collapse
|
38
|
Juhl AD, Lund FW, Jensen MLV, Szomek M, Heegaard CW, Guttmann P, Werner S, McNally J, Schneider G, Kapishnikov S, Wüstner D. Niemann Pick C2 protein enables cholesterol transfer from endo-lysosomes to the plasma membrane for efflux by shedding of extracellular vesicles. Chem Phys Lipids 2021; 235:105047. [PMID: 33422548 DOI: 10.1016/j.chemphyslip.2020.105047] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
The Niemann-Pick C2 protein (NPC2) is a sterol transfer protein in the lumen of late endosomes and lysosomes (LE/LYSs). Absence of functional NPC2 leads to endo-lysosomal buildup of cholesterol and other lipids. How NPC2's known capacity to transport cholesterol between model membranes is linked to its function in living cells is not known. Using quantitative live-cell imaging combined with modeling of the efflux kinetics, we show that NPC2-deficient human fibroblasts can export the cholesterol analog dehydroergosterol (DHE) from LE/LYSs. Internalized NPC2 accelerated sterol efflux extensively, accompanied by reallocation of LE/LYSs containing fluorescent NPC2 and DHE to the cell periphery. Using quantitative fluorescence loss in photobleaching of TopFluor-cholesterol (TF-Chol), we estimate a residence time for a rapidly exchanging sterol pool in LE/LYSs localized in close proximity to the plasma membrane (PM), of less than one min and observed non-vesicular sterol exchange between LE/LYSs and the PM. Excess sterol was released from the PM by shedding of cholesterol-rich vesicles. The ultrastructure of such vesicles was analyzed by combined fluorescence and cryo soft X-ray tomography (SXT), revealing that they can contain lysosomal cargo and intraluminal vesicles. Treating cells with apoprotein A1 and with nuclear receptor liver X-receptor (LXR) agonists to upregulate expression of ABC transporters enhanced cholesterol efflux from the PM, at least partly by accelerating vesicle release. We conclude that NPC2 inside LE/LYSs facilitates non-vesicular sterol exchange with the PM for subsequent sterol efflux to acceptor proteins and for shedding of sterol-rich vesicles from the cell surface.
Collapse
Affiliation(s)
- Alice Dupont Juhl
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Frederik W Lund
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Maria Louise V Jensen
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Maria Szomek
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark
| | - Christian W Heegaard
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000, Aarhus C, Denmark
| | - Peter Guttmann
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - Stephan Werner
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - James McNally
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - Gerd Schneider
- Department X-Ray Microscopy, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, 12489, Berlin, Germany
| | - Sergey Kapishnikov
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, DK-5230, Odense M, Denmark.
| |
Collapse
|
39
|
|
40
|
Analysis of Correlations Between Selected Blood Markers of Liver Function and Milk Composition in Cows During Late Lactation Period. ANNALS OF ANIMAL SCIENCE 2020. [DOI: 10.2478/aoas-2020-0020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Abstract
The knowledge of the existing levels and the interrelationships between various blood and milk parameters is very useful for the analysis and monitoring of homeostasis high-yielding dairy cows. The aim of the study was to evaluate these values and correlations for selected blood markers of liver function aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma-glutamyl transferase (GGT), bilirubin, albumin, glucose, cholesterol) and selected milk parameters: somatic cell count (SCC), colony-forming units (CFU), fat, protein, lactose, dry matter (DM), fat-free dry matter (FDM), and milk production in cows during late lactation period. At the same time blood and milk samples were collected from 11 clinically healthy milking cows in later lactation period. The 11 selected cows were examined once a day for 3 days resulting in 33 sets of blood and milk samples for laboratory and statistical analysis. Significant positive correlations were observed between: ALT and albumin, ALT and cholesterol, GGT and glucose, albumin and cholesterol, CFU and fat, CFU and DM, SCC and protein, fat and DM, protein and FDM, lactose and FDM, GPT and FDM, albumin and protein, albumin and FDM, glucose and fat, as well as significant negative correlations between: AST and ALT, AST and GGT, AST and albumin, CFU and lactose, fat and lactose, fat and FDM, lactose and DM, DM and FDM, AST and SCC, AST and protein, AST and FDM, ALT and fat, ALT and DM, glucose and DM, cholesterol and CFU. The results obtained may be important for detecting of different biochemical pathways and helpful in estimating, predicting or determining trends, the direction of changes in liver functions and assessing the risk of alert levels for liver blood markers, when only daily results of milk parameters are available.
Collapse
|
41
|
Cedó L, Metso J, Santos D, García-León A, Plana N, Sabate-Soler S, Rotllan N, Rivas-Urbina A, Méndez-Lara KA, Tondo M, Girona J, Julve J, Pallarès V, Benitez-Amaro A, Llorente-Cortes V, Pérez A, Gómez-Coronado D, Ruotsalainen AK, Levonen AL, Sanchez-Quesada JL, Masana L, Kovanen PT, Jauhiainen M, Lee-Rueckert M, Blanco-Vaca F, Escolà-Gil JC. LDL Receptor Regulates the Reverse Transport of Macrophage-Derived Unesterified Cholesterol via Concerted Action of the HDL-LDL Axis: Insight From Mouse Models. Circ Res 2020; 127:778-792. [PMID: 32495699 DOI: 10.1161/circresaha.119.316424] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE The HDL (high-density lipoprotein)-mediated stimulation of cellular cholesterol efflux initiates macrophage-specific reverse cholesterol transport (m-RCT), which ends in the fecal excretion of macrophage-derived unesterified cholesterol (UC). Early studies established that LDL (low-density lipoprotein) particles could act as efficient intermediate acceptors of cellular-derived UC, thereby preventing the saturation of HDL particles and facilitating their cholesterol efflux capacity. However, the capacity of LDL to act as a plasma cholesterol reservoir and its potential impact in supporting the m-RCT pathway in vivo both remain unknown. OBJECTIVE We investigated LDL contributions to the m-RCT pathway in hypercholesterolemic mice. METHODS AND RESULTS Macrophage cholesterol efflux induced in vitro by LDL added to the culture media either alone or together with HDL or ex vivo by plasma derived from subjects with familial hypercholesterolemia was assessed. In vivo, m-RCT was evaluated in mouse models of hypercholesterolemia that were naturally deficient in CETP (cholesteryl ester transfer protein) and fed a Western-type diet. LDL induced the efflux of radiolabeled UC from cultured macrophages, and, in the simultaneous presence of HDL, a rapid transfer of the radiolabeled UC from HDL to LDL occurred. However, LDL did not exert a synergistic effect on HDL cholesterol efflux capacity in the familial hypercholesterolemia plasma. The m-RCT rates of the LDLr (LDL receptor)-KO (knockout), LDLr-KO/APOB100, and PCSK9 (proprotein convertase subtilisin/kexin type 9)-overexpressing mice were all significantly reduced relative to the wild-type mice. In contrast, m-RCT remained unchanged in HAPOB100 Tg (human APOB100 transgenic) mice with fully functional LDLr, despite increased levels of plasma APO (apolipoprotein)-B-containing lipoproteins. CONCLUSIONS Hepatic LDLr plays a critical role in the flow of macrophage-derived UC to feces, while the plasma increase of APOB-containing lipoproteins is unable to stimulate m-RCT. The results indicate that, besides the major HDL-dependent m-RCT pathway via SR-BI (scavenger receptor class B type 1) to the liver, a CETP-independent m-RCT path exists, in which LDL mediates the transfer of cholesterol from macrophages to feces. Graphical Abstract: A graphical abstract is available for this article.
Collapse
Affiliation(s)
- Lídia Cedó
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| | - Jari Metso
- Minerva Foundation Institute for Medical Research and National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum, Helsinki, Finland (J.M., M.J.)
| | - David Santos
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| | - Annabel García-León
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Núria Plana
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.).,Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain (N.P., J.G., L.M.)
| | - Sonia Sabate-Soler
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Noemí Rotllan
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Andrea Rivas-Urbina
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Karen A Méndez-Lara
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Mireia Tondo
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Josefa Girona
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain (N.P., J.G., L.M.)
| | - Josep Julve
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| | - Victor Pallarès
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.)
| | - Aleyda Benitez-Amaro
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, Institut de Recerca Josep Carreras, Barcelona, Spain (V.P.); Biomedical Research Institute Sant Pau (IIB Sant Pau), Institute of Biomedical Research of Barcelona-Spanish National Research Council (A.B.-A., V.L.-C.)
| | - Vicenta Llorente-Cortes
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, Institut de Recerca Josep Carreras, Barcelona, Spain (V.P.); Biomedical Research Institute Sant Pau (IIB Sant Pau), Institute of Biomedical Research of Barcelona-Spanish National Research Council (A.B.-A., V.L.-C.).,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain (V.L.-C.)
| | - Antonio Pérez
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| | - Diego Gómez-Coronado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain (D.G.-C.).,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain (D.G.-C.)
| | - Anna-Kaisa Ruotsalainen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio (A.-K.R., A.-L.L.)
| | - Anna-Liisa Levonen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio (A.-K.R., A.-L.L.)
| | - José Luis Sanchez-Quesada
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| | - Luís Masana
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.).,Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain (N.P., J.G., L.M.)
| | - Petri T Kovanen
- and Wihuri Research Institute, Helsinki, Finland (P.T.K., M.L.-R.)
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research and National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum, Helsinki, Finland (J.M., M.J.)
| | | | - Francisco Blanco-Vaca
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| | - Joan Carles Escolà-Gil
- From the Institut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain (L.C., D.S., A.G.-L., S.S.-S., N.R., A.R.-U., K.A.M.-L., M.T., J.J., V.P., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Spain (A.R.-U., K.A.M.-L., J.J., A.P., J.L.S.-Q., F.B.-V., J.C.E.-G.).,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain (L.C., D.S., N.P., J.J., A.P., J.L.S.-Q., L.M., F.B.-V., J.C.E.-G.)
| |
Collapse
|
42
|
Cosset FL, Mialon C, Boson B, Granier C, Denolly S. HCV Interplay with Lipoproteins: Inside or Outside the Cells? Viruses 2020; 12:v12040434. [PMID: 32290553 PMCID: PMC7232430 DOI: 10.3390/v12040434] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/05/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a major public health issue leading to chronic liver diseases. HCV particles are unique owing to their particular lipid composition, namely the incorporation of neutral lipids and apolipoproteins. The mechanism of association between HCV virion components and these lipoproteins factors remains poorly understood as well as its impact in subsequent steps of the viral life cycle, such as entry into cells. It was proposed that the lipoprotein biogenesis pathway is involved in HCV morphogenesis; yet, recent evidence indicated that HCV particles can mature and evolve biochemically in the extracellular medium after egress. In addition, several viral, cellular and blood components have been shown to influence and regulate this specific association. Finally, this specific structure and composition of HCV particles was found to influence entry into cells as well as their stability and sensitivity to neutralizing antibodies. Due to its specific particle composition, studying the association of HCV particles with lipoproteins remains an important goal towards the rational design of a protective vaccine.
Collapse
|
43
|
In vitro conditions for performance evaluation of products for intravascular administration: Developing appropriate test media using Amphotericin B as a model drug. Eur J Pharm Sci 2020; 143:105174. [DOI: 10.1016/j.ejps.2019.105174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/15/2019] [Accepted: 12/02/2019] [Indexed: 11/24/2022]
|
44
|
Changes over time in serum albumin levels predict outcomes following percutaneous coronary intervention. J Cardiol 2019; 75:381-386. [PMID: 31547947 DOI: 10.1016/j.jjcc.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/12/2019] [Accepted: 08/26/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Serum albumin (SA) is a powerful prognostic marker in patients with cardiovascular diseases. OBJECTIVES To evaluate the prognostic significance of changes in SA levels among patients that underwent percutaneous coronary intervention (PCI). METHODS Patients who underwent PCI in a tertiary medical center throughout 2004-2017 were considered for the study. Patients with missing SA values within the year before or 5 years after the PCI were excluded from the study as well as those for whom SA values were obtained throughout an acute infection. Changes in albumin were represented by Δalbumin: Albumin before PCI (closest) minus lowest albumin within the 5 years following the PCI. Δalbumin was categorized into 4 categories as follows: ≤0 g/dL, 0-0.5 g/dL, 0.5-1 g/dL, ≥1 g/dL. Primary outcome was major adverse cardiac events (MACE) that included all-cause mortality, non-fatal myocardial infarction, target vessel re-vascularization, and coronary artery bypass surgery. RESULTS The study included 5449 out of 21,763 (25%) patients, mean age 66.8 ± 12 years, 26% women. Patients with greater Δalbumin were older with higher prevalence of most cardiovascular risk factors and comorbidity. The follow-up period was 7 years (median), with cumulative MACE rate of 49.5%. MACE rates and the rates of all the MACE components increased gradually with the increase in Δalbumin. Multivariate adjusted analysis showed that Δalbumin is an independent predictor of long-term MACE following PCI [HR = 1.4 (95%CI:1.3-1.6), HR = 2.2 (95%CI:1.9-2.5), HR = 3.8 (95%CI:3.3-4.35) for Δalbumin of 0-0.5 g/dL, 0.5-1 g/dL, ≥1 g/dL, respectivelyp < 0.001 for all]. CONCLUSIONS A decrease in albumin levels following PCI is an independent prognostic marker of worse long-term outcomes.
Collapse
|
45
|
Abstract
Introduction: High-density lipoprotein (HDL) particles are heterogeneous and their proteome is complex and distinct from HDL cholesterol. However, it is largely unknown whether HDL proteins are associated with cardiovascular protection. Areas covered: HDL isolation techniques and proteomic analyses are reviewed. A list of HDL proteins reported in 37 different studies was compiled and the effects of different isolation techniques on proteins attributed to HDL are discussed. Mass spectrometric techniques used for HDL analysis and the need for precise and robust methods for quantification of HDL proteins are discussed. Expert opinion: Proteins associated with HDL have the potential to be used as biomarkers and/or help to understand HDL functionality. To achieve this, large cohorts must be studied using precise quantification methods. Key factors in HDL proteome quantification are the isolation methodology and the mass spectrometry technique employed. Isolation methodology affects what proteins are identified in HDL and the specificity of association with HDL particles needs to be addressed. Shotgun proteomics yields imprecise quantification, but the majority of HDL studies relied on this approach. Few recent studies used targeted tandem mass spectrometry to quantify HDL proteins, and it is imperative that future studies focus on the application of these precise techniques.
Collapse
Affiliation(s)
- Graziella Eliza Ronsein
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo , São Paulo , Brazil
| | - Tomáš Vaisar
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington , Seattle , WA , USA
| |
Collapse
|
46
|
Neufeld EB, Sato M, Gordon SM, Durbhakula V, Francone N, Aponte A, Yilmaz G, Sviridov D, Sampson M, Tang J, Pryor M, Remaley AT. ApoA-I-Mediated Lipoprotein Remodeling Monitored with a Fluorescent Phospholipid. BIOLOGY 2019; 8:E53. [PMID: 31336888 PMCID: PMC6784057 DOI: 10.3390/biology8030053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
We describe simple, sensitive and robust methods to monitor lipoprotein remodeling and cholesterol and apolipoprotein exchange, using fluorescent Lissamine Rhodamine B head-group tagged phosphatidylethanolamine (*PE) as a lipoprotein reference marker. Fluorescent Bodipy cholesterol (*Chol) and *PE directly incorporated into whole plasma lipoproteins in proportion to lipoprotein cholesterol and phospholipid mass, respectively. *Chol, but not *PE, passively exchanged between isolated plasma lipoproteins. Fluorescent apoA-I (*apoA-I) specifically bound to high-density lipoprotein (HDL) and remodeled *PE- and *Chol-labeled synthetic lipoprotein-X multilamellar vesicles (MLV) into a pre-β HDL-like particle containing *PE, *Chol, and *apoA-I. Fluorescent MLV-derived *PE specifically incorporated into plasma HDL, whereas MLV-derived *Chol incorporation into plasma lipoproteins was similar to direct *Chol incorporation, consistent with apoA-I-mediated remodeling of fluorescent MLV to HDL with concomitant exchange of *Chol between lipoproteins. Based on these findings, we developed a model system to study lipid transfer by depositing fluorescent *PE and *Chol-labeled on calcium silicate hydrate crystals, forming dense lipid-coated donor particles that are readily separated from acceptor lipoprotein particles by low-speed centrifugation. Transfer of *PE from donor particles to mouse plasma lipoproteins was shown to be HDL-specific and apoA-I-dependent. Transfer of donor particle *PE and *Chol to HDL in whole human plasma was highly correlated. Taken together, these studies suggest that cell-free *PE efflux monitors apoA-I functionality.
Collapse
Affiliation(s)
- Edward B Neufeld
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Masaki Sato
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott M Gordon
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vinay Durbhakula
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Francone
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angel Aponte
- Proteomics Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gizem Yilmaz
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Denis Sviridov
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maureen Sampson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jingrong Tang
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Milton Pryor
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Lai SJ, Ohkawa R, Horiuchi Y, Kubota T, Tozuka M. Red blood cells participate in reverse cholesterol transport by mediating cholesterol efflux of high-density lipoprotein and apolipoprotein A-I from THP-1 macrophages. Biol Chem 2019; 400:1593-1602. [DOI: 10.1515/hsz-2019-0244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/04/2019] [Indexed: 11/15/2022]
Abstract
Abstract
High-density lipoprotein (HDL) plays a main role in reverse cholesterol transport (RCT), one of the most important functions for preventing atherosclerosis. Recent reports have shown that red blood cells (RBCs) can be associated with RCT, an interaction facilitated by albumin. However, the RCT function of RBCs has not been thoroughly elucidated. In this study, the RCT function of RBCs was assessed using cholesterol efflux capacity (CEC) assays, in which [3H]-labeled cholesterol-loaded human acute monocytic leukemia (THP-1) macrophages were incubated with RBCs as a cholesterol acceptor in the presence or absence of HDL or its main component protein apolipoprotein A-I (apoA-I). The CEC of RBCs was found to be dose dependent, enabling uptake of cholesterol from THP-1 macrophages through apoA-I and HDL, and directly from apoA-I and HDL in medium without the presence THP-1 macrophages. Moreover, RBCs could exchange cholesterol with HDL in a bidirectional manner but could only exchange cholesterol with apoA-I in a single direction. Although albumin promoted the movement of cholesterol, synergistic effects were not observed for both apoA-I and HDL, in contrast to previous findings. These results strongly suggested that RBCs may play important roles in RCT by mediating cholesterol efflux as temporary cholesterol storage.
Collapse
Affiliation(s)
- Shao-Jui Lai
- Analytical Laboratory Chemistry, Field of Applied Laboratory Science, Graduate School of Health Care Science , Tokyo Medical and Dental University , 1-5-45 Yushima, Bunkyo-ku , Tokyo 113-8519 , Japan
| | - Ryunosuke Ohkawa
- Analytical Laboratory Chemistry, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , 1-5-45 Yushima, Bunkyo-ku , Tokyo 113-8519 , Japan
| | - Yuna Horiuchi
- Analytical Laboratory Chemistry, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , 1-5-45 Yushima, Bunkyo-ku , Tokyo 113-8519 , Japan
| | - Tetsuo Kubota
- Immunopathology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , 1-5-45 Yushima, Bunkyo-ku , Tokyo 113-8519 , Japan
| | - Minoru Tozuka
- Analytical Laboratory Chemistry, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , 1-5-45 Yushima, Bunkyo-ku , Tokyo 113-8519 , Japan
- Life Science Research Center , Nagano Children’s Hospital , 3100 Toyoshina , Azumino 399-8288 , Japan
| |
Collapse
|
48
|
Denolly S, Granier C, Fontaine N, Pozzetto B, Bourlet T, Guérin M, Cosset FL. A serum protein factor mediates maturation and apoB-association of HCV particles in the extracellular milieu. J Hepatol 2019; 70:626-638. [PMID: 30553840 DOI: 10.1016/j.jhep.2018.11.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 11/15/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS In the sera of infected patients, hepatitis C virus (HCV) particles display heterogeneous forms with low-buoyant densities (<1.08), underscoring their lipidation via association with apoB-containing lipoproteins, which was proposed to occur during assembly or secretion from infected hepatocytes. However, the mechanisms inducing this association remain poorly-defined and most cell culture grown HCV (HCVcc) particles exhibit higher density (>1.08) and poor/no association with apoB. We aimed to elucidate the mechanisms of lipidation and to produce HCVcc particles resembling those in infected sera. METHODS We produced HCVcc particles of Jc1 or H77 strains from Huh-7.5 hepatoma cells cultured in standard conditions (10%-fetal calf serum) vs. in serum-free or human serum conditions before comparing their density profiles to patient-derived virus. We also characterized wild-type and Jc1/H77 hypervariable region 1 (HVR1)-swapped mutant HCVcc particles produced in serum-free media and incubated with different serum types or with purified lipoproteins. RESULTS Compared to serum-free or fetal calf serum conditions, production with human serum redistributed most HCVcc infectious particles to low density (<1.08) or very-low density (<1.04) ranges. In addition, short-time incubation with human serum was sufficient to shift HCVcc physical particles to low-density fractions, in time- and dose-dependent manners, which increased their specific infectivity, promoted apoB-association and induced neutralization-resistance. Moreover, compared to Jc1, we detected higher levels of H77 HCVcc infectious particles in very-low-density fractions, which could unambiguously be attributed to strain-specific features of the HVR1 sequence. Finally, all 3 lipoprotein classes, i.e., very-low-density, low-density and high-density lipoproteins, could synergistically induce low-density shift of HCV particles; yet, this required additional non-lipid serum factor(s) that include albumin. CONCLUSIONS The association of HCV particles with lipids may occur in the extracellular milieu. The lipidation level depends on serum composition as well as on HVR1-specific properties. These simple culture conditions allow production of infectious HCV particles resembling those of chronically-infected patients. LAY SUMMARY Hepatitis C virus (HCV) particles may associate with apoB and acquire neutral lipids after exiting cells, giving them low-buoyant density. The hypervariable region 1 (HVR1) is a majorviral determinant of E2 that controls this process. Besides lipoproteins, specific serum factors including albumin promote extracellular maturation of HCV virions. HCV particle production in vitro, with media of defined serum conditions, enables production of infectious particles resembling those of chronically infected patients.
Collapse
Affiliation(s)
- Solène Denolly
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, F-69007 Lyon, France
| | - Christelle Granier
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, F-69007 Lyon, France
| | - Nelly Fontaine
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, F-69007 Lyon, France
| | - Bruno Pozzetto
- GIMAP, EA 3064, Faculté de Médecine, Université de Saint-Etienne, Univ Lyon, F-42023 Saint Etienne, France
| | - Thomas Bourlet
- GIMAP, EA 3064, Faculté de Médecine, Université de Saint-Etienne, Univ Lyon, F-42023 Saint Etienne, France
| | - Maryse Guérin
- Inserm, Sorbonne-Université, Research Unit of Cardiovascular, Metabolism and Nutrition Diseases UMR_S1166-ICAN, Paris F-75013, France
| | - François-Loïc Cosset
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, F-69007 Lyon, France.
| |
Collapse
|
49
|
Vázquez-Oliva G, Zamora A, Ramos R, Subirana I, Grau M, Dégano IR, Muñoz D, Fitó M, Elosua R, Marrugat J. Analysis of Plasma Albumin, Vitamin D, and Apolipoproteins A and B as Predictive Coronary Risk Biomarkers in the REGICOR Study. ACTA ACUST UNITED AC 2018; 71:910-916. [PMID: 29764762 DOI: 10.1016/j.rec.2018.01.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/08/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION AND OBJECTIVES New biomarkers could improve the predictive capacity of classic risk functions. The aims of this study were to determine the association between circulating levels of apolipoprotein A1 (apoA1), apolipoprotein B (apoB), albumin, and 25-OH-vitamin D and coronary events and to analyze whether these biomarkers improve the predictive capacity of the Framingham-REGICOR risk function. METHODS A case-cohort study was designed. From an initial cohort of 5404 individuals aged 35 to 74 years with a 5-year follow-up, all the participants who had a coronary event (n = 117) and a random group of the cohort (subcohort; n = 667) were selected. Finally, 105 cases and 651 individuals representative of the cohort with an available biological sample were included. The events of interest were angina, fatal and nonfatal myocardial infarction and coronary deaths. RESULTS Case participants were older, had a higher proportion of men and cardiovascular risk factors, and showed higher levels of apoB and lower levels of apoA1, apoA1/apoB ratio, 25-OH-vitamin D and albumin than the subcohort. In multivariate analyses, plasma albumin concentration was the only biomarker independently associated with coronary events (HR, 0.73; P = .002). The inclusion of albumin in the risk function properly reclassified a significant proportion of individuals, especially in the intermediate risk group (net reclassification improvement, 32.3; P = .048). CONCLUSIONS Plasma albumin levels are inversely associated with coronary risk and improve the predictive capacity of classic risk functions.
Collapse
Affiliation(s)
- Gabriel Vázquez-Oliva
- Departamento de Cardiología, Fundación Althaia, Manresa, Barcelona, Spain; Facultad de Medicina, Universidad de Girona, Girona, Spain
| | - Alberto Zamora
- Facultad de Medicina, Universidad de Girona, Girona, Spain; Unidad de Riesgo Vascular, Hospital de Blanes, Corporación de Salud del Maresme y la Selva, Blanes, Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain
| | - Rafel Ramos
- Facultad de Medicina, Universidad de Girona, Girona, Spain; Instituto Universitario de Investigación en Atención Primaria (IDIAP) Jordi Gol, Grupo de Investigación ISV, Unidad de Investigación en Atención Primaria, Girona, Spain; Atención Primaria, Instituto Catalán de Salud (ICS), Girona, Spain
| | - Isaac Subirana
- Grupo de Epidemiología y Genética Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - María Grau
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain; Grupo de Epidemiología y Genética Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain
| | - Irene R Dégano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain; Grupo de Epidemiología y Genética Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain; Universidad de Vic-Central de Cataluña (UVic-UCC), Vic, Barcelona, Spain
| | - Daniel Muñoz
- Grupo de Nutrición y Riesgo Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain
| | - Montserrat Fitó
- Grupo de Nutrición y Riesgo Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Barcelona, Spain
| | - Roberto Elosua
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain; Grupo de Epidemiología y Genética Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain; Universidad de Vic-Central de Cataluña (UVic-UCC), Vic, Barcelona, Spain
| | - Jaume Marrugat
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain; Grupo de Epidemiología y Genética Cardiovascular, Grupo del Estudio REGICOR (REgistre GIroní del COR), IMIM (Instituto Hospital del Mar de Investigaciones Médicas), Barcelona, Spain.
| |
Collapse
|
50
|
Vázquez-Oliva G, Zamora A, Ramos R, Subirana I, Grau M, Dégano IR, Muñoz D, Fitó M, Elosua R, Marrugat J. Valor predictivo de la albúmina plasmática, la vitamina D y las apolipoproteínas A y B como biomarcadores de riesgo coronario en el estudio REGICOR. Rev Esp Cardiol 2018. [DOI: 10.1016/j.recesp.2018.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|