1
|
Chunta S, Khongwichit S, Watanasin P, Lieberzeit PA, Amatatongchai M. Design of hybrid aptamer-molecularly imprinted polymer nanoparticles for selective binding of oxidized low-density lipoprotein in an ELISA-mimic system. Talanta 2025; 287:127605. [PMID: 39832470 DOI: 10.1016/j.talanta.2025.127605] [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: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Oxidized low-density lipoprotein (oxLDL) is the leading cause of atherosclerosis and cardiovascular disease development. An enzyme-linked immunosorbent assay (ELISA)-mimic system for sensitive and specific oxLDL determination was developed using selective aptamer-molecularly imprinted polymer nanoparticles (AP-MIP NP) coupled with an immunology-based colorimetric assay. The AP-MIP NP were synthesized using solid-phase molecular imprinting by incorporating aptamers into the MIP NP cavities. This resulted in AP-MIP NP with diameters of 108 ± 28 nm. For AP-MIP NP-ELISA-mimic assay development, the surface of a microplate was coated with the novel AP-MIP NP capture receptors at a concentration of 0.1 mg mL-1 to capture oxLDL. The reaction time between AP-MIP NP and oxLDL was 20 min. Horseradish peroxidase conjugated anti-oxLDL polyclonal antibody at a concentration of 0.6 μg mL-1 was used as the detection antibody, with a linear response ranging from 24.72 to 1,600 μg dL-1. The recovery accuracy was 89-106 %. Within-run precision was 3.3-6.7 % of the coefficient of variation, while between-day precision was 3.8-7.1 %. The AP-MIP NP-coated wells were stored at room temperature for one month without a loss of binding ability, retaining over 91 % binding ability after three regeneration cycles. Human serum diluted 1:10 and analyzed by the AP-MIP NP-ELISA-mimic assay showed high correlation with conventional ELISA (R2 = 0.9779). This assay achieved rapid results within 95 min, compared to ELISA at 195 min. The high binding ability and selectivity of the AP-MIP NP shows promise as a selective material against oxLDL for the ELISA-mimic system and other applications.
Collapse
Affiliation(s)
- Suticha Chunta
- Department of Clinical Chemistry, Faculty of Medical Technology, Prince of Songkla University, Hatyai, Songkla, 90110, Thailand.
| | - Soemwit Khongwichit
- Department of Clinical Chemistry, Faculty of Medical Technology, Prince of Songkla University, Hatyai, Songkla, 90110, Thailand; Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Panwadee Watanasin
- Department of Analytical Chemistry, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Peter A Lieberzeit
- Department of Physical Chemistry, Faculty for Chemistry, University of Vienna, Vienna, 1090, Austria
| | - Maliwan Amatatongchai
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| |
Collapse
|
2
|
Berndsen ZT, Cassidy CK. The structure of apolipoprotein B100 from human low-density lipoprotein. Nature 2025; 638:836-843. [PMID: 39662503 PMCID: PMC11839476 DOI: 10.1038/s41586-024-08467-w] [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: 01/31/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024]
Abstract
Low-density lipoprotein (LDL) has a central role in lipid and cholesterol metabolism and is a key agent in the development and progression of atherosclerosis, the leading cause of mortality worldwide1,2. Apolipoprotein B100 (apoB100), one of the largest proteins in the genome, is the primary structural and functional component of LDL, yet its size and complex lipid associations have posed major challenges for structural studies3. Here we present the structure of apoB100 resolved to subnanometre resolution in most regions using an integrative approach of cryo-electron microscopy, AlphaFold24 and molecular-dynamics-based refinement5. The structure consists of a large globular N-terminal domain and an approximately 61-nm-long continuous amphipathic β-sheet that wraps around the LDL particle like a belt. Distributed quasi-symmetrically across the two sides of the β-belt are nine strategically located interstrand inserts that extend across the lipid surface to provide additional structural support through a network of long-range interactions. We further compare our structure to a comprehensive list of more than 200 intramolecular cross-links and find close agreement between the two. These results suggest a mechanism for how the various domains of apoB100 act in concert to maintain LDL shape and cohesion across a range of particle sizes. More generally, they advance our fundamental understanding of LDL synthesis, form and function, and will help to accelerate the design of potential therapeutics.
Collapse
Affiliation(s)
| | - C Keith Cassidy
- Department of Physics, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
3
|
Santos HO, Penha-Silva N. Revisiting the concepts of de novo lipogenesis to understand the conversion of carbohydrates into fats: Stop overvaluing and extrapolating the renowned phrase "fat burns in the flame of carbohydrate". Nutrition 2025; 130:112617. [PMID: 39566326 DOI: 10.1016/j.nut.2024.112617] [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: 11/05/2023] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Carbohydrates can be converted into fatty acids via de novo lipogenesis (DNL). Although DNL is considered inefficient, these endogenous fatty acids contribute substantially to the esterification pathway in adipose tissue, together with fatty acids of feeding. This article revisited the concepts of DNL and aimed to discuss the clinical magnitude of carbohydrate overfeeding and fat mass accumulation. Although fat storage resulting from fat intake is more favorable for fat mass accrual than carbohydrates due to molecule structure and metabolism (e.g., oxidation and thermic effect), carbohydrates can substantially participate in lipogenesis and esterification under excess carbohydrate intake over time. Regarding only monosaccharide overfeeding, glucose and fructose favor the subcutaneous and visceral adipose tissue, respectively. While fructose and sucrose are considered villains in nonalcoholic fatty liver disease, energy surplus from carbohydrates, regardless of sources, can be considered an underlying cause of obesity. Interestingly, some degree of DNL in adipocytes may be favorable to mitigate a high deposition of fatty acids in the liver, conferring a physiological role. Although "fat burns in the flame of carbohydrate" is a praiseworthy phrase that has helped describe basic concepts in biochemistry for many decades, it appears to be overvalued and extrapolated even nowadays. DNL cannot be neglected. It is time to consider DNL an efficient biochemical process in health and disease.
Collapse
Affiliation(s)
- Heitor O Santos
- School of Medicine, Uberlândia Federal University, Uberlândia, MG, Brazil.
| | - Nilson Penha-Silva
- Institute of Biotechnology, Uberlândia Federal University, Uberlândia, MG, Brazil
| |
Collapse
|
4
|
Khongwichit S, Swangphon P, Nualla-ong A, Prompat N, Amatatongchai M, Lieberzeit PA, Chunta S. Reduced Uptake of Oxidized Low-Density Lipoprotein by Macrophages Using Multiple Aptamer Combinations. ACS APPLIED BIO MATERIALS 2025; 8:457-474. [PMID: 39762152 PMCID: PMC11752521 DOI: 10.1021/acsabm.4c01432] [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: 09/30/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/21/2025]
Abstract
The accumulation of oxidized low-density lipoprotein (oxLDL) in macrophages leads to the formation of foam cells and atherosclerosis development. Reducing the uptake of oxLDL in macrophages decreases the incidence and progression of atherosclerosis. Four distinct single-strand DNA sequences, namely, AP07, AP11, AP25, and AP29, were selected that demonstrated specific binding to distinct regions of oxidized apolipoprotein B100 (apoB100; the protein component of oxLDL) with low HDOCK scores. These four DNA sequences were combined to generate aptamers that selectively bound to labeled Dil-oxLDL, and were subsequently added to murine RAW 264.7 macrophages to test their inhibitory effects using fluorescence spectrometry. The four combined aptamers at 10 μM reduced oxLDL uptake by 79 ± 4% compared to that of the untreated aptamer group. Flow cytometry data demonstrated that macrophages treated with aptamers reached only 32.6% of the Dil-oxLDL signal, a 50% reduction in fluorescence emission relative to that of the untreated group (64.4% Dil-oxLDL signal). Binding the four combined aptamers to the oxLDL surface disrupted the interaction between oxLDL and CD36 via cyclic voltammetry, effectively decreasing the level of uptake of oxLDL by macrophages. Results suggested that these aptamers could be used as alternative compounds to prevent the formation of foam cells, hence providing antiatherosclerosis activity.
Collapse
Affiliation(s)
- Soemwit Khongwichit
- Faculty
of Medical Technology, Prince of Songkla University, Songkhla 90110, Thailand
- Division
of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Piyawut Swangphon
- Faculty
of Medical Technology, Prince of Songkla University, Songkhla 90110, Thailand
| | - Aekkaraj Nualla-ong
- Division
of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
- Center
for Genomics and Bioinformatic Research, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
- Medical
of Technology Service Center, Faculty of
Medical Technology, Prince of Songkla University, Songkhla 90110, Thailand
| | - Napat Prompat
- Faculty
of Medical Technology, Prince of Songkla University, Songkhla 90110, Thailand
- Medical
of Technology Service Center, Faculty of
Medical Technology, Prince of Songkla University, Songkhla 90110, Thailand
| | - Maliwan Amatatongchai
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
| | - Peter A. Lieberzeit
- Department
of Physical Chemistry, Faculty for Chemistry, University of Vienna, Vienna 1090, Austria
| | - Suticha Chunta
- Faculty
of Medical Technology, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
5
|
Chambers KL, Myerscough MR, Watson MG, Byrne HM. Blood Lipoproteins Shape the Phenotype and Lipid Content of Early Atherosclerotic Lesion Macrophages: A Dual-Structured Mathematical Model. Bull Math Biol 2024; 86:112. [PMID: 39093509 PMCID: PMC11297092 DOI: 10.1007/s11538-024-01342-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Macrophages in atherosclerotic lesions exhibit a spectrum of behaviours or phenotypes. The phenotypic distribution of monocyte-derived macrophages (MDMs), its correlation with MDM lipid content, and relation to blood lipoprotein densities are not well understood. Of particular interest is the balance between low density lipoproteins (LDL) and high density lipoproteins (HDL), which carry bad and good cholesterol respectively. To address these issues, we have developed a mathematical model for early atherosclerosis in which the MDM population is structured by phenotype and lipid content. The model admits a simpler, closed subsystem whose analysis shows how lesion composition becomes more pathological as the blood density of LDL increases relative to the HDL capacity. We use asymptotic analysis to derive a power-law relationship between MDM phenotype and lipid content at steady-state. This relationship enables us to understand why, for example, lipid-laden MDMs have a more inflammatory phenotype than lipid-poor MDMs when blood LDL lipid density greatly exceeds HDL capacity. We show further that the MDM phenotype distribution always attains a local maximum, while the lipid content distribution may be unimodal, adopt a quasi-uniform profile or decrease monotonically. Pathological lesions exhibit a local maximum in both the phenotype and lipid content MDM distributions, with the maximum at an inflammatory phenotype and near the lipid content capacity respectively. These results illustrate how macrophage heterogeneity arises in early atherosclerosis and provide a framework for future model validation through comparison with single-cell RNA sequencing data.
Collapse
Affiliation(s)
- Keith L Chambers
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK.
| | - Mary R Myerscough
- School of Mathematics and Statistics, University of Sydney, Carslaw Building, Eastern Avenue, Camperdown, Sydney, NSW, 2006, Australia
| | - Michael G Watson
- School of Mathematics and Statistics, University of New South Wales, Anita B. Lawrence Centre, University Mall, UNSW, Kensington, Sydney, NSW, 2052, Australia
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Headington, Oxford, Oxfordshire, OX3 7DQ, UK
| |
Collapse
|
6
|
Gilbert MAG, Fatima N, Jenkins J, O'Sullivan TJ, Schertel A, Halfon Y, Wilkinson M, Morrema THJ, Geibel M, Read RJ, Ranson NA, Radford SE, Hoozemans JJM, Frank RAW. CryoET of β-amyloid and tau within postmortem Alzheimer's disease brain. Nature 2024; 631:913-919. [PMID: 38987603 PMCID: PMC11269202 DOI: 10.1038/s41586-024-07680-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 06/06/2024] [Indexed: 07/12/2024]
Abstract
A defining pathological feature of most neurodegenerative diseases is the assembly of proteins into amyloid that form disease-specific structures1. In Alzheimer's disease, this is characterized by the deposition of β-amyloid and tau with disease-specific conformations. The in situ structure of amyloid in the human brain is unknown. Here, using cryo-fluorescence microscopy-targeted cryo-sectioning, cryo-focused ion beam-scanning electron microscopy lift-out and cryo-electron tomography, we determined in-tissue architectures of β-amyloid and tau pathology in a postmortem Alzheimer's disease donor brain. β-amyloid plaques contained a mixture of fibrils, some of which were branched, and protofilaments, arranged in parallel arrays and lattice-like structures. Extracellular vesicles and cuboidal particles defined the non-amyloid constituents of β-amyloid plaques. By contrast, tau inclusions formed parallel clusters of unbranched filaments. Subtomogram averaging a cluster of 136 tau filaments in a single tomogram revealed the polypeptide backbone conformation and filament polarity orientation of paired helical filaments within tissue. Filaments within most clusters were similar to each other, but were different between clusters, showing amyloid heterogeneity that is spatially organized by subcellular location. The in situ structural approaches outlined here for human donor tissues have applications to a broad range of neurodegenerative diseases.
Collapse
Affiliation(s)
- Madeleine A G Gilbert
- Astbury Centre for Structural Molecular Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Nayab Fatima
- Astbury Centre for Structural Molecular Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Joshua Jenkins
- Astbury Centre for Structural Molecular Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Thomas J O'Sullivan
- Astbury Biostructure Laboratory CryoEM facility, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Andreas Schertel
- ZEISS Microscopy Customer Center Europe, Carl Zeiss Microscopy GmbH, Oberkochen, Germany
| | - Yehuda Halfon
- Astbury Biostructure Laboratory CryoEM facility, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Martin Wilkinson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Tjado H J Morrema
- Department of Pathology, Unit Neuropathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mirjam Geibel
- ZEISS Microscopy Customer Center Europe, Carl Zeiss Microscopy GmbH, Oberkochen, Germany
| | - Randy J Read
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Neil A Ranson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Jeroen J M Hoozemans
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - René A W Frank
- Astbury Centre for Structural Molecular Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
7
|
Zimmerman AJ, de Oliveira GP, Su X, Wood J, Fu Z, Pinckney B, Tigges J, Ghiran I, Ivanov AR. Multimode chromatography-based techniques for high purity isolation of extracellular vesicles from human blood plasma. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e147. [PMID: 38751711 PMCID: PMC11080799 DOI: 10.1002/jex2.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 03/08/2024] [Indexed: 05/18/2024]
Abstract
Extracellular vesicles (EVs) play a pivotal role in various biological pathways, such as immune responses and the progression of diseases, including cancer. However, it is challenging to isolate EVs at high purity from blood plasma and other biofluids due to their low abundance compared to more predominant biomolecular species such as lipoprotein particles and free protein complexes. Ultracentrifugation-based EV isolation, the current gold standard technique, cannot overcome this challenge due to the similar biophysical characteristics of such species. We developed several novel approaches to enrich EVs from plasma while depleting contaminating molecular species using multimode chromatography-based strategies. On average, we identified 716 ± 68 and 1054 ± 35 protein groups in EV isolates from 100 µL of plasma using multimode chromatography- and ultracentrifugation-based techniques, respectively. The developed methods resulted in similar EV isolates purity, providing significant advantages in simplicity, throughput, scalability, and applicability for various downstream analytical and potential clinical applications.
Collapse
Affiliation(s)
- Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Getulio Pereira de Oliveira
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Xianyi Su
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Jacqueline Wood
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Zhengxin Fu
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Brandy Pinckney
- Nano Flow Core Facility, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - John Tigges
- Nano Flow Core Facility, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Ionita Ghiran
- Department of Anesthesia, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| |
Collapse
|
8
|
Nakamura Y, Kulkarni NN, Takahashi T, Alimohamadi H, Dokoshi T, Liu E, Shia M, Numata T, Luo EW, Gombart AF, Yang X, Secrest P, Gordts PL, Tsimikas S, Wong GC, Gallo RL. Increased LL37 in psoriasis and other inflammatory disorders promotes LDL uptake and atherosclerosis. J Clin Invest 2024; 134:e172578. [PMID: 38194294 PMCID: PMC10904043 DOI: 10.1172/jci172578] [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: 05/25/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Patients with chronic inflammatory disorders such as psoriasis have an increased risk of cardiovascular disease and elevated levels of LL37, a cathelicidin host defense peptide that has both antimicrobial and proinflammatory properties. To explore whether LL37 could contribute to the risk of heart disease, we examined its effects on lipoprotein metabolism and show that LL37 enhanced LDL uptake in macrophages through the LDL receptor (LDLR), scavenger receptor class B member 1 (SR-B1), and CD36. This interaction led to increased cytosolic cholesterol in macrophages and changes in expression of lipid metabolism genes consistent with increased cholesterol uptake. Structure-function analysis and synchrotron small-angle x-ray scattering showed structural determinants of the LL37-LDL complex that underlie its ability to bind its receptors and promote uptake. This function of LDL uptake is unique to cathelicidins from humans and some primates and was not observed with cathelicidins from mice or rabbits. Notably, Apoe-/- mice expressing LL37 developed larger atheroma plaques than did control mice, and a positive correlation between plasma LL37 and oxidized phospholipid on apolipoprotein B (OxPL-apoB) levels was observed in individuals with cardiovascular disease. These findings provide evidence that LDL uptake can be increased via interaction with LL37 and may explain the increased risk of cardiovascular disease associated with chronic inflammatory disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Adrian F. Gombart
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | | | - Patrick Secrest
- Department of Medicine, Division of Endocrinology and Metabolism, and
| | - Philip L.S.M. Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, and
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | | | - Gerard C.L. Wong
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | | |
Collapse
|
9
|
Lam M, Kuo SY, Reis S, Gestwicki JE, Silva MC, Haggarty SJ. Cholesterol Dysregulation Drives Seed-Dependent Tau Aggregation in Patient Stem Cell-Derived Models of Tauopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571147. [PMID: 38168389 PMCID: PMC10759997 DOI: 10.1101/2023.12.11.571147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the progressive misfolding and accumulation of pathological tau protein in focal regions of the brain, leading to insidious neurodegeneration. Abnormalities in cholesterol metabolism and homeostasis have also been implicated in various neurodegenerative diseases. However, the connection between cholesterol dysregulation and tau pathology remains largely unknown. To model and measure the impact of cholesterol dysregulation on tau, we utilized a combination of in vitro and ex vivo tau aggregation assays using an engineered tau biosensor cell line and human induced pluripotent stem cell (iPSC)-derived neuronal cultures from an individual harboring an autosomal dominant P301L tau mutation and from a healthy control. We demonstrate that excess cholesterol esters lead to an increased rate of tau aggregation in vitro and an increase in seed-dependent insoluble tau aggregates detected in the biosensor line. We observed a strong correlation between cholesterol ester concentration and the presence of high-molecular-weight, oligomeric tau species. Importantly, in tauopathy patient iPSC-derived neurons harboring a P301L tau mutation with endogenous forms of misfolded tau, we show that acute dysregulation of cholesterol homeostasis through acute exposure to human plasma-purified cholesterol esters formed by the linkage of fatty acids to the hydroxyl group of cholesterol leads to the rapid accumulation of phosphorylated tau. Conversely, treatment with the same cholesterol esters pool did not lead to subsequent accumulation of phosphorylated tau in control iPSC-derived neurons. Finally, treatment with a heterobifunctional, small-molecule degrader designed to selectively engage and catalyze the ubiquitination and proteasomal degradation of aberrant tau species prevented cholesterol ester-induced aggregation of tau in the biosensor cell line in a Cereblon E3 ligase-dependent manner. Degrader treatment also restored the resiliency of tauopathy patient-derived neurons towards cholesterol ester-induced tau aggregation phenotypes. Taken together, our study supports a key role of cholesterol dysregulation in tau aggregation. Moreover, it provides further pre-clinical validation of the therapeutic strategy of targeted protein degradation with heterobifunctional tau degraders for blocking tau seeding.
Collapse
|
10
|
Cisse A, Desfosses A, Stainer S, Kandiah E, Traore DAK, Bezault A, Schachner-Nedherer AL, Leitinger G, Hoerl G, Hinterdorfer P, Gutsche I, Prassl R, Peters J, Kornmueller K. Targeting structural flexibility in low density lipoprotein by integrating cryo-electron microscopy and high-speed atomic force microscopy. Int J Biol Macromol 2023; 252:126345. [PMID: 37619685 DOI: 10.1016/j.ijbiomac.2023.126345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/26/2023]
Abstract
Low-density lipoprotein (LDL) plays a crucial role in cholesterol metabolism. Responsible for cholesterol transport from the liver to the organs, LDL accumulation in the arteries is a primary cause of cardiovascular diseases, such as atherosclerosis. This work focuses on the fundamental question of the LDL molecular structure, as well as the topology and molecular motions of apolipoprotein B-100 (apo B-100), which is addressed by single-particle cryo-electron microscopy (cryo-EM) and high-speed atomic force microscopy (HS-AFM). Our results suggest a revised model of the LDL core organization with respect to the cholesterol ester (CE) arrangement. In addition, a high-density region close to the flattened poles could be identified, likely enriched in free cholesterol. The most remarkable new details are two protrusions on the LDL surface, attributed to the protein apo B-100. HS-AFM adds the dimension of time and reveals for the first time a highly dynamic direct description of LDL, where we could follow large domain fluctuations of the protrusions in real time. To tackle the inherent flexibility and heterogeneity of LDL, the cryo-EM maps are further assessed by 3D variability analysis. Our study gives a detailed explanation how to approach the intrinsic flexibility of a complex system comprising lipids and protein.
Collapse
Affiliation(s)
- Aline Cisse
- Université Grenoble Alpes, CNRS, LiPhy, Grenoble, France; Institut Laue-Langevin, Grenoble, France
| | - Ambroise Desfosses
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
| | - Sarah Stainer
- Department of Experimental Applied Biophysics, Johannes Kepler University Linz, Linz, Austria
| | | | - Daouda A K Traore
- Institut Laue-Langevin, Grenoble, France; Faculté de Pharmacie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali; Faculty of Natural Sciences, School of Life Sciences, Keele University, Staffordshire, UK
| | - Armel Bezault
- Institut Européen de Chimie et Biologie, UAR3033/US001, Université de Bordeaux, CNRS, INSERM 2, Pessac, France; Structural Image Analysis Unit, Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR3528, Paris, France
| | - Anna-Laurence Schachner-Nedherer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics Division, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Gerd Hoerl
- Otto Loewi Research Center, Physiological Chemistry, Medical University of Graz, Graz, Austria
| | - Peter Hinterdorfer
- Department of Experimental Applied Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Irina Gutsche
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
| | - Ruth Prassl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics Division, Medical University of Graz, Graz, Austria
| | - Judith Peters
- Université Grenoble Alpes, CNRS, LiPhy, Grenoble, France; Institut Laue-Langevin, Grenoble, France; Institut Universitaire de France, France.
| | - Karin Kornmueller
- Institut Laue-Langevin, Grenoble, France; Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics Division, Medical University of Graz, Graz, Austria.
| |
Collapse
|
11
|
Wang J, Tian L, Wu K, Wang C, Zhu C. Reconstituting Low-Density Lipoprotein with NIR-Absorbing Organic Photothermal Agents for Targeted Killing of Cancer Cells. Macromol Rapid Commun 2023; 44:e2300395. [PMID: 37566746 DOI: 10.1002/marc.202300395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/05/2023] [Indexed: 08/13/2023]
Abstract
Photothermal therapy (PTT) systems typically do not possess intrinsic tumor-targeting capability, resulting in indiscriminate thermal damage to both cancer and normal cells. Herein, a low-density lipoprotein (LDL)-based nanosystem (denoted as MTTQ@LDL) is reported for targeted photothermal killing of cancer cells. Such a nanosystem is fabricated by reconstituting the lipophilic core of LDL with an organic photothermal agent MTTQ. The reconstitution process improves the supramolecular photothermal effects of MTTQ assemblies, which contributes to the significantly enhanced photothermal conversion efficiency (41.3% vs. 16.2%). MTTQ@LDL can actively target LDL receptor-overexpressed cancer cells via receptor-mediated endocytosis, enabling the selective killing of cancer cells over normal cells (98% vs. 7%) post-NIR irradiation. Reconstituted LDL can serve as a promising platform for targeted delivery of functional materials, holding great promise in tumor eradication in vivo.
Collapse
Affiliation(s)
- Jiaxin Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Liang Tian
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Kaiyu Wu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chao Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chunlei Zhu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
12
|
Sellem L, Eichelmann F, Jackson KG, Wittenbecher C, Schulze MB, Lovegrove JA. Replacement of dietary saturated with unsaturated fatty acids is associated with beneficial effects on lipidome metabolites: a secondary analysis of a randomized trial. Am J Clin Nutr 2023:S0002-9165(23)46314-9. [PMID: 37062359 DOI: 10.1016/j.ajcnut.2023.03.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND The effects of replacing dietary saturated fatty acids (SFAs) with monounsaturated fatty acids (MUFAs) and/or polyunsaturated fatty acids (PUFAs) on the plasma lipidome in relation to the cardiometabolic disease (CMD) risk are poorly understood. OBJECTIVES We aimed to assess the impact of substituting dietary SFAs with unsaturated fatty acids (UFAs) on the plasma lipidome and examine the relationship between lipid metabolites modulated by diet and CMD risk. METHODS Plasma fatty acid (FA) concentrations among 16 lipid classes (within-class FAs) were measured in a subgroup from the Dietary Intervention and VAScular function (DIVAS) parallel randomized controlled trial (n = 113/195), which consisted of three 16-wk diets enriched in SFAs (target SFA:MUFA:n-6PUFA ratio = 17:11:4% total energy [TE]), MUFAs (9:19:4% TE), or a MUFA/PUFA mixture (9:13:10% TE). Similar lipidomics analyses were conducted in the European investigation into Cancer and Nutrition (EPIC)-Potsdam prospective cohort study (specific case/cohorts: n = 775/1886 for type 2 diabetes [T2D], n = 551/1671 for cardiovascular disease [CVD]). Multiple linear regression and multivariable Cox models identified within-class FAs sensitive to replacement of dietary SFA with UFA in DIVAS and their association with CMD risk in EPIC-Potsdam. Elastic-net regression models identified within-class FAs associated with changes in CMD risk markers post-DIVAS interventions. RESULTS DIVAS high-UFA interventions reduced plasma within-class FAs associated with a higher CVD risk in EPIC-Potsdam, especially SFA-containing glycerolipids and sphingolipids (e.g., diacylglycerol (20:0) z-score = -1.08; SE = 0.17; P value < 10-8), whereas they increased those inversely associated with CVD risk. The results on T2D were less clear. Specific sphingolipids and phospholipids were associated with changes in markers of endothelial function and ambulatory blood pressure, whereas higher low-density lipoprotein cholesterol concentrations were characterized by higher plasma glycerolipids containing lauric and stearic acids. CONCLUSIONS These results suggest a mediating role of plasma lipid metabolites in the association between dietary fat and CMD risk. Future research combining interventional and observational findings will further our understanding of the role of dietary fat in CMD etiology. This trial was registered in ClinicalTrials.gov as NCT01478958.
Collapse
Affiliation(s)
- Laury Sellem
- Hugh Sinclair Unit of Human Nutrition, and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Harry Nursten Building, Reading, UK
| | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Kim G Jackson
- Hugh Sinclair Unit of Human Nutrition, and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Harry Nursten Building, Reading, UK
| | - Clemens Wittenbecher
- Division of Food Science and Nutrition, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Harry Nursten Building, Reading, UK.
| |
Collapse
|
13
|
Berg AR, Petrole RF, Li H, Sorokin AV, Gonzalez-Cantero A, Playford MP, Mehta NN, Teague HL. Cholesterol efflux capacity is associated with lipoprotein size and vascular health in mild to moderate psoriasis. Front Cardiovasc Med 2023; 10:1041457. [PMID: 36891247 PMCID: PMC9986595 DOI: 10.3389/fcvm.2023.1041457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/24/2023] [Indexed: 02/22/2023] Open
Abstract
Background and objective Psoriasis is a systemic inflammatory condition with poor cholesterol transport measured by cholesterol efflux capacity (CEC) that is associated with a heightened risk of cardiovascular disease (CVD). In psoriasis patients, we sought to characterize the lipoprotein profile by size using a novel nuclear magnetic resonance algorithm in patients with low CEC compared to normal CEC. Methods Lipoprotein profile was assessed using the novel nuclear magnetic resonance LipoProfile-4 deconvolution algorithm. Aortic vascular inflammation (VI) and non-calcified burden (NCB) were characterized via positron emission tomography-computed tomography and coronary computed tomography angiography. To understand the relationship between lipoprotein size and markers of subclinical atherosclerosis, linear regression models controlling for confounders were constructed. Results Psoriasis patients with low CEC had higher more severe psoriasis (p = 0.04), VI (p = 0.04) and NCB (p = 0.001), concomitant with smaller high-density lipoprotein (HDL) (p < 0.001) and low-density lipoprotein (LDL) particles (p < 0.001). In adjusted models HDL size (β = -0.19; p = 0.02) and LDL size (β = -0.31; p < 0.001) associated with VI and NCB. Lastly, HDL size strongly associated with LDL size in fully adjusted models (β = -0.27; p < 0.001). Conclusion These findings demonstrate that in psoriasis, low CEC associates with a lipoprotein profile comprised of smaller HDL and LDL particles which correlates with vascular health and may be driving early onset atherogenesis. Further, these results demonstrate a relationship between HDL and LDL size and provide novel insights into the complexities of HDL and LDL as biomarkers of vascular health.
Collapse
Affiliation(s)
- Alexander R Berg
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| | - Rylee F Petrole
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| | - Haiou Li
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| | | | - Alvaro Gonzalez-Cantero
- Dermatology Service, Hospital Universitario Ramón y Cajal, Medicine Department, Faculty of Medicine, Universidad de Alcalá, IRYCIS, Madrid, Spain.,Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| | - Heather L Teague
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
14
|
Wong SK, Ramli FF, Ali A, Ibrahim N‘I. Genetics of Cholesterol-Related Genes in Metabolic Syndrome: A Review of Current Evidence. Biomedicines 2022; 10:biomedicines10123239. [PMID: 36551995 PMCID: PMC9775320 DOI: 10.3390/biomedicines10123239] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome (MetS) refers to a cluster of metabolic dysregulations, which include insulin resistance, obesity, atherogenic dyslipidemia and hypertension. The complex pathogenesis of MetS encompasses the interplay between environmental and genetic factors. Environmental factors such as excessive nutrients and sedentary lifestyle are modifiable and could be improved by lifestyle modification. However, genetic susceptibility to MetS, a non-modifiable factor, has attracted the attention of researchers, which could act as the basis for future diagnosis, prognosis, and therapy for MetS. Several cholesterol-related genes associated with each characteristic of MetS have been identified, such as apolipoprotein, lipoprotein lipase (LPL), cholesteryl ester transfer protein (CETP) and adiponectin. This review aims to summarize the genetic information of cholesterol-related genes in MetS, which may potentially serve as biomarkers for early prevention and management of MetS.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Fitri Fareez Ramli
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
- Clinical Psychopharmacology Research Unit, Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Adli Ali
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
- Correspondence: ; Tel.: +60-39145-9545
| |
Collapse
|
15
|
Hou S, Hasnat M, Chen Z, Liu Y, Faran Ashraf Baig MM, Liu F, Chen Z. Application Perspectives of Nanomedicine in Cancer Treatment. Front Pharmacol 2022; 13:909526. [PMID: 35860027 PMCID: PMC9291274 DOI: 10.3389/fphar.2022.909526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is a disease that seriously threatens human health. Based on the improvement of traditional treatment methods and the development of new treatment modes, the pattern of cancer treatment is constantly being optimized. Nanomedicine plays an important role in these evolving tumor treatment modalities. In this article, we outline the applications of nanomedicine in three important tumor-related fields: chemotherapy, gene therapy, and immunotherapy. According to the current common problems, such as poor targeting of first-line chemotherapy drugs, easy destruction of nucleic acid drugs, and common immune-related adverse events in immunotherapy, we discuss how nanomedicine can be combined with these treatment modalities, provide typical examples, and summarize the advantages brought by the application of nanomedicine.
Collapse
Affiliation(s)
- Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Muhammad Hasnat
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Ziwei Chen
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Yinong Liu
- Hospital Laboratory of Nangjing Lishui People’s Hospital, Nangjing, China
| | - Mirza Muhammad Faran Ashraf Baig
- Laboratory of Biomedical Engineering for Novel Bio-functional, and Pharmaceutical Nanomaterials, Prince Philip Dental Hospital, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fuhe Liu
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
- *Correspondence: Zelong Chen, ; Fuhe Liu,
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Province Engineering Research Center of Artificial Intelligence and Internet of Things Wise Medical, Zhengzhou, China
- *Correspondence: Zelong Chen, ; Fuhe Liu,
| |
Collapse
|
16
|
Nimer R, Kamel G, Obeidat MA, Dahabiyeh LA. Investigating the molecular structure of plasma in type 2 diabetes mellitus and diabetic nephropathy by synchrotron Fourier-transform infrared microspectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 264:120259. [PMID: 34388428 DOI: 10.1016/j.saa.2021.120259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 06/13/2023]
Abstract
Diabetes mellitus (DM) is associated with a high incidence of morbidity and mortality which, in many cases, is derived from the progressive kidney dysfunction due to diabetic nephropathy (DN). In this study, synchrotron-Fourier-transform infrared (SR-FTIR) microspectroscopy was used to identify molecular changes in the lipid and protein regions in the plasma of patients with different stages of DN (mild, moderate, severe and end-stage), and patients with type 2 diabetes mellitus (T2DM) without DN. Our results revealed different conformational changes in the proteins secondary structure between DN stages, and between DN and T2DM groups illustrated by peak shifts and intensity alterations. End-stage DN showed the highest CH2/CH3 ratio and intensity of the carbonyl group in protein-carbonyl region compared to other DN stages indicating high level of unsaturation and lipid peroxidation and oxidation conditions. Moreover, end-stage DN group was characterized by a decrease in amide I and amide II absorption signals which reflected a sign of hypoalbuminemia. When compared to T2DM, DN group demonstrated a higher oxidation state as confirmed via the high intensity of the carbonyl group and the high level of malondialdehyde. The current study highlights the promising role of SR-FTIR microspectroscopy as a new sensitive analytical approach that can be used to provide better understanding of the pathophysiology of DN, and guide the development of new preventive therapies and treatments.
Collapse
Affiliation(s)
- Refat Nimer
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, 22110 Irbid, Jordan.
| | - Gihan Kamel
- SESAME Synchrotron (Synchrotron-light for Experimental Science and Applications in the Middle East), 19252 Allan, Jordan; Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt
| | - Motaz A Obeidat
- Department of Internal Medicine, Nephrology Division, Jordan University of Science and Technology, Irbid, Jordan
| | - Lina A Dahabiyeh
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, 11942 Amman, Jordan.
| |
Collapse
|
17
|
Rubina KA, Semina EV, Kalinina NI, Sysoeva VY, Balatskiy AV, Tkachuk VA. Revisiting the multiple roles of T-cadherin in health and disease. Eur J Cell Biol 2021; 100:151183. [PMID: 34798557 DOI: 10.1016/j.ejcb.2021.151183] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 01/02/2023] Open
Abstract
As a non-canonical member of cadherin superfamily, T-cadherin was initially described as a molecule involved in homophilic recognition in the nervous and vascular systems. The ensuing decades clearly demonstrated that T-cadherin is a remarkably multifunctional molecule. It was validated as a bona fide receptor for both: LDL exerting adverse atherogenic action and adiponectin mediating many protective metabolic and cardiovascular effects. Motivated by the latest progress and accumulated data unmasking important roles of T-cadherin in blood vessel function and tissue regeneration, here we revisit the original function of T-cadherin as a guidance receptor for the growing axons and blood vessels, consider the recent data on T-cadherin-induced exosomes' biogenesis and their role in myocardial regeneration and revascularization. The review expands upon T-cadherin contribution to mesenchymal stem/stromal cell compartment in adipose tissue. We also dwell upon T-cadherin polymorphisms (SNP) and their possible therapeutic applications. Furthermore, we scrutinize the molecular hub of insulin and adiponectin receptors (AdipoR1 and AdipoR2) conveying signals to their downstream targets in quest for defining a putative place of T-cadherin in this molecular circuitry.
Collapse
Affiliation(s)
- K A Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia.
| | - E V Semina
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - N I Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V Yu Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - A V Balatskiy
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V A Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| |
Collapse
|
18
|
Iqbal H, Yang T, Li T, Zhang M, Ke H, Ding D, Deng Y, Chen H. Serum protein-based nanoparticles for cancer diagnosis and treatment. J Control Release 2020; 329:997-1022. [PMID: 33091526 DOI: 10.1016/j.jconrel.2020.10.030] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022]
Abstract
Serum protein as naturally essential biomacromolecules has recently emerged as a versatile carrier for diagnostic and therapeutic drug delivery for cancer nanomedicine with superior biocompatibility, improved pharmacokinetics and enhanced targeting capacity. A variety of serum proteins have been utilized for drug delivery, mainly including albumin, ferritin/apoferritin, transferrin, low-density lipoprotein, high-density lipoprotein and hemoglobin. As evidenced by the success of paclitaxel-bound albumin nanoparticles (AbraxaneTM), serum protein-based nanoparticles have gained attractive attentions for precise biological design and potential clinical application. In this review, we summarize the general design strategies, targeting mechanisms and recent development of serum protein-based nanoparticles in the field of cancer nanomedicine. Moreover, we also concisely specify the current challenges to be addressed for a bright future of serum protein-based nanomedicines.
Collapse
Affiliation(s)
- Haroon Iqbal
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ting Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Miya Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Hengte Ke
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Dawei Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| | - Huabing Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
19
|
Jakubauskas D, Jansen M, Lyngsø J, Cheng Y, Pedersen JS, Cárdenas M. Toward reliable low-density lipoprotein ultrastructure prediction in clinical conditions: A small-angle X-ray scattering study on individuals with normal and high triglyceride serum levels. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102318. [PMID: 33091569 DOI: 10.1016/j.nano.2020.102318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/23/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Atherosclerosis is the main killer in the west and therefore a major health challenge today. Total serum cholesterol and lipoprotein concentrations, used as clinical markers, fail to predict the majority of cases, especially between the risk scale extremes, due to the high complexity in lipoprotein structure and composition. In particular, low-density lipoprotein (LDL) plays a key role in atherosclerosis development, with LDL size being a parameter considered for determining the risk for cardiovascular diseases. Determining LDL size and structural parameters is challenging to address experimentally under physiological-like conditions. This article describes the biochemistry and ultrastructure of normolipidemic and hypertriglyceridemic LDL fractions and subfractions using small-angle X-ray scattering. Our results conclude that LDL particles of hypertriglyceridemic compared to healthy individuals 1) have lower LDL core melting temperature, 2) have lower cholesteryl ester ordering in their core, 3) are smaller, rounder and more spherical below melting temperature, and 4) their protein-containing shell is thinner above melting temperature.
Collapse
Affiliation(s)
- Dainius Jakubauskas
- Biofilms - Research center for Biointerfaces, Dept. of Biomedical Science, Faculty of Health and Society, Malmo University, Malmo, Sweden.
| | - Martin Jansen
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Centre, University of Freiburg, Freiburg im Breisgau, Germany.
| | - Jeppe Lyngsø
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark.
| | - Yuanji Cheng
- Department of Materials Science and Applied Mathematics, Faculty of Technology and Society, Malmo University, Malmo, Sweden.
| | - Jan Skov Pedersen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark.
| | - Marité Cárdenas
- Biofilms - Research center for Biointerfaces, Dept. of Biomedical Science, Faculty of Health and Society, Malmo University, Malmo, Sweden.
| |
Collapse
|
20
|
Direct electrochemiluminescent immunosensing for an early indication of coronary heart disease using dual biomarkers. Anal Chim Acta 2020; 1110:82-89. [DOI: 10.1016/j.aca.2020.03.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/07/2020] [Accepted: 03/11/2020] [Indexed: 11/18/2022]
|
21
|
Starich MR, Tang J, Remaley AT, Tjandra N. Squeezing lipids: NMR characterization of lipoprotein particles under pressure. Chem Phys Lipids 2020; 228:104874. [PMID: 31978380 PMCID: PMC7232691 DOI: 10.1016/j.chemphyslip.2020.104874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 11/25/2022]
Abstract
Determining the particle size and number of lipoprotein components found in blood plasma (HDL, LDL and VLDL) has become an important clinical tool in diagnosing risk of cardiovascular disease. Proton (1H) NMR spectroscopy methods to quantify lipoprotein particle subclasses have been advancing since NMR lineshape analysis of plasma samples was first proposed in the 1990's. NMR methods, including a more recent DOSY-based diffusion spectroscopy test, provide the foundation for the advanced lipoprotein tests, including Lipoprotein® and Liposcale® analyses available for clinical use to determine particle size and number. At the time of this submission, no NMR studies exist which explore physical parameters of individual lipoprotein fractions when they are deformed by pressure. This study reports 1H NMR frequency shifts and T2* measurements for the broad methyl peak attributed to terminal methyls (cholesteryl positions 26, 27 and terminal acyl methyl groups) in three primary lipoprotein fractions as a function of hydraulic pressure. This terminal CH3 resonance shifted linearly upfield as a function of pressure for HDL and VLDL (observed slopes of -0.014 Hz/bar). The LDL terminal CH3 resonance shows segmented behavior, with a shallow slope between 0-900 bar (-0.008 hz/bar) and a slope similar to HDL and VDL across the range from 1000 to 2400 bar (slope -0.016 Hz/bar). 1H T2* values measured for VLDL and HDL dropped linearly with increasing pressure. 1H T2* values for LDL demonstrated segmented behavior as a function of pressure. The unique behavior observed for LDL terminal CH3 frequency and 1H T2* trends suggests an approximate pressure at which phase transition occurs.
Collapse
Affiliation(s)
- Mary R Starich
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Jingrong Tang
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Nico Tjandra
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
22
|
Different spatiotemporal organization of GPI-anchored T-cadherin in response to low-density lipoprotein and adiponectin. Biochim Biophys Acta Gen Subj 2019; 1863:129414. [DOI: 10.1016/j.bbagen.2019.129414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 07/23/2019] [Accepted: 08/07/2019] [Indexed: 01/10/2023]
|
23
|
Jutkova A, Chorvat D, Miskovsky P, Jancura D, Datta S. Encapsulation of anticancer drug curcumin and co-loading with photosensitizer hypericin into lipoproteins investigated by fluorescence resonance energy transfer. Int J Pharm 2019; 564:369-378. [DOI: 10.1016/j.ijpharm.2019.04.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/17/2019] [Accepted: 04/20/2019] [Indexed: 01/19/2023]
|
24
|
Liu J, Wu H, Huang C, Lei D, Zhang M, Xie W, Li J, Ren G. Optimized Negative-Staining Protocol for Lipid-Protein Interactions Investigated by Electron Microscopy. Methods Mol Biol 2019; 2003:163-173. [PMID: 31218618 PMCID: PMC6817366 DOI: 10.1007/978-1-4939-9512-7_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A large number of proteins are capable of inserting themselves into lipids, and interacting with membranes, such as transmembrane proteins and apolipoproteins. Insights into the lipid-protein interactions are important in understanding biological processes, and the structure of proteins at the lipid binding stage can help identify their roles and critical functions. Previously, such structural determination was challenging to obtain because the traditional methods, such as X-ray crystallography, are unable to capture the conformational and compositional heterogeneity of protein-lipid complexes. Electron microscopy (EM) is an alternative approach to determining protein structures and visualizing lipid-protein interactions directly, and negative-staining (OpNS), a subset of EM techniques, is a rapid, frequently used qualitative approach. The concern, however, is that current NS protocols often generate artifacts with lipid-related proteins, such as rouleaux formation from lipoproteins. To overcome this artifact formation, Ren and his colleagues have refined early NS protocols, and developed an optimized NS protocol that validated by comparing images of lipoproteins from cryo-electron microscopy (cryo-EM). This optimized NS protocol produces "near native-state" particle images and high contrast images of the protein in its native lipid-binding state, which can be used to create higher-quality three-dimensional (3D) reconstruction by single-particle analysis and electron tomography (e.g. IPET). This optimized protocol is thus a promising hands-on approach for examining the structure of proteins at their lipid-binding status.
Collapse
Affiliation(s)
- Jianfang Liu
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Hao Wu
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department of Computer Science, College of Information Science and Technology, Beijing Normal University, Beijing, China
| | - Changyu Huang
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Dongsheng Lei
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Meng Zhang
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Wei Xie
- State Key Laboratory for Biocontrol, School of Life Sciences, The Sun Yat-Sen University, Guangzhou, Guangdong, China
- Center for Cellular and Structural Biology, The Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jinping Li
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Gang Ren
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
25
|
Lehofer B, Golub M, Kornmueller K, Kriechbaum M, Martinez N, Nagy G, Kohlbrecher J, Amenitsch H, Peters J, Prassl R. High Hydrostatic Pressure Induces a Lipid Phase Transition and Molecular Rearrangements in Low-Density Lipoprotein Nanoparticles. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2018; 35:1800149. [PMID: 30283212 PMCID: PMC6166783 DOI: 10.1002/ppsc.201800149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Low-density lipoproteins (LDL) are natural lipid transporter in human plasma whose chemically modified forms contribute to the progression of atherosclerosis and cardiovascular diseases accounting for a vast majority of deaths in westernized civilizations. For the development of new treatment strategies, it is important to have a detailed picture of LDL nanoparticles on a molecular basis. Through the combination of X-ray and neutron small-angle scattering (SAS) techniques with high hydrostatic pressure (HHP) this study describes structural features of normolipidemic, triglyceride-rich and oxidized forms of LDL. Due to the different scattering contrasts for X-rays and neutrons, information on the effects of HHP on the internal structure determined by lipid rearrangements and changes in particle shape becomes accessible. Independent pressure and temperature variations provoke a phase transition in the lipid core domain. With increasing pressure an inter-related anisotropic deformation and flattening of the particle are induced. All LDL nanoparticles maintain their structural integrity even at 3000 bar and show a reversible response toward pressure variations. The present work depicts the complementarity of pressure and temperature as independent thermodynamic parameters and introduces HHP as a tool to study molecular assembling and interaction processes in distinct lipoprotein particles in a nondestructive manner.
Collapse
Affiliation(s)
- Bernhard Lehofer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Maksym Golub
- Institut Laue-Langevin, 71 avenue des Martyrs, 38044 Grenoble, France; Univ. Grenoble Alpes, CNRS + CEA, IBS, 38000 Grenoble, France
| | - Karin Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Manfred Kriechbaum
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Nicolas Martinez
- Institut Laue-Langevin, 71 avenue des Martyrs, 38044 Grenoble, France; Univ. Grenoble Alpes, CNRS + CEA, IBS, 38000 Grenoble, France
| | - Gergely Nagy
- Paul Scherrer Institut, 5232 Villigen, Switzerland; Wigner Research Centre for Physics, 1121 Budapest, Hungary; European Spallation Source ERIC, 22363 Lund, Sweden
| | | | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Judith Peters
- Institut Laue-Langevin, 71 avenue des Martyrs, 38044 Grenoble, France; Univ. Grenoble Alpes, CNRS, LiPhy, 38000 Grenoble, France
| | - Ruth Prassl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| |
Collapse
|
26
|
Jayaprakash K, Demirel I, Khalaf H, Bengtsson T. Porphyromonas gingivalis-induced inflammatory responses in THP1 cells are altered by native and modified low-density lipoproteins in a strain-dependent manner. APMIS 2018; 126:667-677. [DOI: 10.1111/apm.12860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/19/2018] [Indexed: 01/08/2023]
Affiliation(s)
| | - Isak Demirel
- Department of Medical Sciences; Örebro University; Örebro Sweden
| | - Hazem Khalaf
- Department of Medical Sciences; Örebro University; Örebro Sweden
| | | |
Collapse
|
27
|
Datta S, Hovan A, Jutková A, Kruglik SG, Jancura D, Miskovsky P, Bánó G. Phosphorescence Kinetics of Singlet Oxygen Produced by Photosensitization in Spherical Nanoparticles. Part II. The Case of Hypericin-Loaded Low-Density Lipoprotein Particles. J Phys Chem B 2018; 122:5154-5160. [PMID: 29709185 DOI: 10.1021/acs.jpcb.8b00659] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The phosphorescence kinetics of singlet oxygen produced by photosensitized hypericin (Hyp) molecules inside low-density lipoprotein (LDL) particles was studied experimentally and by means of numerical and analytical modeling. The phosphorescence signal was measured after short laser pulse irradiation of aqueous Hyp/LDL solutions. The Hyp triplet state lifetime determined by a laser flash-photolysis measurement was 5.3 × 10-6 s. The numerical and the analytical model described in part I of the present work (DOI: 10.1021/acs.jpcb.8b00658) were used to analyze the observed phosphorescence kinetics of singlet oxygen. It was shown that singlet oxygen diffuses out of LDL particles on a time scale shorter than 0.1 μs. The total (integrated) concentration of singlet oxygen inside LDL is more than an order of magnitude smaller than the total singlet oxygen concentration in the solvent. The time course of singlet oxygen concentrations inside and outside the particles was calculated using simplified representations of the LDL internal structure. The experimental phosphorescence data were fitted by a linear combination of these concentrations using the emission factor E (the ratio of the radiative singlet oxygen depopulation rate constants inside and outside LDL) as a fitting parameter. The emission factor was determined to be E = 6.7 ± 2.5. Control measurements were carried out by adding sodium azide, a strong singlet oxygen quencher, to the solution.
Collapse
Affiliation(s)
| | | | | | - Sergei G Kruglik
- Laboratoire Jean Perrin, Sorbonne Universités, UPMC Univ. Paris 6, CNRS UMR 8237 , 4 place Jussieu , 75005 Paris , France
| | | | | | | |
Collapse
|
28
|
Robson AL, Dastoor PC, Flynn J, Palmer W, Martin A, Smith DW, Woldu A, Hua S. Advantages and Limitations of Current Imaging Techniques for Characterizing Liposome Morphology. Front Pharmacol 2018; 9:80. [PMID: 29467660 PMCID: PMC5808202 DOI: 10.3389/fphar.2018.00080] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/24/2018] [Indexed: 11/13/2022] Open
Abstract
There are currently a number of imaging techniques available for evaluating the morphology of liposomes and other nanoparticles, with each having its own advantages and disadvantages that should be considered when interpreting data. Controlling and validating the morphology of nanoparticles is of key importance for the effective clinical translation of liposomal formulations. There are a number of physical characteristics of liposomes that determine their in vivo behavior, including size, surface characteristics, lamellarity, and homogeneity. Despite the great importance of the morphology of nanoparticles, it is generally not well-characterized and is difficult to control. Appropriate imaging techniques provide important details regarding the morphological characteristics of nanoparticles, and should be used in conjunction with other methods to assess physicochemical parameters. In this review, we will discuss the advantages and limitations of available imaging techniques used to evaluate liposomal formulations.
Collapse
Affiliation(s)
- Annie-Louise Robson
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Paul C Dastoor
- Centre for Organic Electronics, University of Newcastle, Callaghan, NSW, Australia
| | - Jamie Flynn
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - William Palmer
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Antony Martin
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Doug W Smith
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Ameha Woldu
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Susan Hua
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
29
|
Praphakar RA, Jeyaraj M, Mehnath S, Higuchi A, Ponnamma D, Sadasivuni KK, Rajan M. A pH-sensitive guar gum-grafted-lysine-β-cyclodextrin drug carrier for the controlled release of 5-flourouracil into cancer cells. J Mater Chem B 2018; 6:1519-1530. [DOI: 10.1039/c7tb02551c] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The physiological environment is a crucial factor in biomedical systems, which can be regulated with relative ease both in vitro and in vivo.
Collapse
Affiliation(s)
- Rajendran Amarnath Praphakar
- Biomaterials in Medicinal Chemistry Laboratory
- Department of Natural Products Chemistry
- School of Chemistry
- Madurai Kamaraj University
- Madurai-625021
| | - Murugan Jeyaraj
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai
- India
| | - Sivaraj Mehnath
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai
- India
| | - Akon Higuchi
- Department of Chemical and Materials Engineering
- National Central University
- Jhong-Li
- Taiwan
| | | | | | - Mariappan Rajan
- Biomaterials in Medicinal Chemistry Laboratory
- Department of Natural Products Chemistry
- School of Chemistry
- Madurai Kamaraj University
- Madurai-625021
| |
Collapse
|
30
|
Jena P, Roxbury D, Galassi TV, Akkari L, Horoszko CP, Iaea DB, Budhathoki-Uprety J, Pipalia N, Haka AS, Harvey JD, Mittal J, Maxfield FR, Joyce JA, Heller DA. A Carbon Nanotube Optical Reporter Maps Endolysosomal Lipid Flux. ACS NANO 2017; 11:10689-10703. [PMID: 28898055 PMCID: PMC5707631 DOI: 10.1021/acsnano.7b04743] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/31/2017] [Indexed: 05/18/2023]
Abstract
Lipid accumulation within the lumen of endolysosomal vesicles is observed in various pathologies including atherosclerosis, liver disease, neurological disorders, lysosomal storage disorders, and cancer. Current methods cannot measure lipid flux specifically within the lysosomal lumen of live cells. We developed an optical reporter, composed of a photoluminescent carbon nanotube of a single chirality, that responds to lipid accumulation via modulation of the nanotube's optical band gap. The engineered nanomaterial, composed of short, single-stranded DNA and a single nanotube chirality, localizes exclusively to the lumen of endolysosomal organelles without adversely affecting cell viability or proliferation or organelle morphology, integrity, or function. The emission wavelength of the reporter can be spatially resolved from within the endolysosomal lumen to generate quantitative maps of lipid content in live cells. Endolysosomal lipid accumulation in cell lines, an example of drug-induced phospholipidosis, was observed for multiple drugs in macrophages, and measurements of patient-derived Niemann-Pick type C fibroblasts identified lipid accumulation and phenotypic reversal of this lysosomal storage disease. Single-cell measurements using the reporter discerned subcellular differences in equilibrium lipid content, illuminating significant intracellular heterogeneity among endolysosomal organelles of differentiating bone-marrow-derived monocytes. Single-cell kinetics of lipoprotein-derived cholesterol accumulation within macrophages revealed rates that differed among cells by an order of magnitude. This carbon nanotube optical reporter of endolysosomal lipid content in live cells confers additional capabilities for drug development processes and the investigation of lipid-linked diseases.
Collapse
Affiliation(s)
- Prakrit
V. Jena
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Daniel Roxbury
- Department
of Chemical Engineering, University of Rhode
Island, Kingston, Rhode Island 02881, United States
| | - Thomas V. Galassi
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
- Weill
Cornell Medicine, New York, New York 10065, United States
| | - Leila Akkari
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
- Division
of Tumor Biology & Immunology, The Netherlands
Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Christopher P. Horoszko
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
- Weill
Cornell Medicine, New York, New York 10065, United States
| | - David B. Iaea
- Weill
Cornell Medicine, New York, New York 10065, United States
| | | | - Nina Pipalia
- Weill
Cornell Medicine, New York, New York 10065, United States
| | - Abigail S. Haka
- Weill
Cornell Medicine, New York, New York 10065, United States
| | - Jackson D. Harvey
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
- Weill
Cornell Medicine, New York, New York 10065, United States
| | - Jeetain Mittal
- Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | | | - Johanna A. Joyce
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
- Weill
Cornell Medicine, New York, New York 10065, United States
- Ludwig Center
for Cancer Research, University of Lausanne, Lausanne CH 1066, Switzerland
| | - Daniel A. Heller
- Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
- Weill
Cornell Medicine, New York, New York 10065, United States
| |
Collapse
|
31
|
Imaging and lipidomics methods for lipid analysis in metabolic and cardiovascular disease. J Dev Orig Health Dis 2017; 8:566-574. [PMID: 28697812 DOI: 10.1017/s2040174417000496] [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] [Indexed: 12/11/2022]
Abstract
Cardiometabolic diseases exhibit changes in lipid biology, which is important as lipids have critical roles in membrane architecture, signalling, hormone synthesis, homoeostasis and metabolism. However, Developmental Origins of Health and Disease studies of cardiometabolic disease rarely include analysis of lipids. This short review highlights some examples of lipid pathology and then explores the technology available for analysing lipids, focussing on the need to develop imaging modalities for intracellular lipids. Analytical methods for studying interactions between the complex endocrine and intracellular signalling pathways that regulate lipid metabolism have been critical in expanding our understanding of how cardiometabolic diseases develop in association with obesity and dietary factors. Biochemical methods can be used to generate detailed lipid profiles to establish links between lifestyle factors and metabolic signalling pathways and determine how changes in specific lipid subtypes in plasma and homogenized tissue are associated with disease progression. New imaging modalities enable the specific visualization of intracellular lipid traffic and distribution in situ. These techniques provide a dynamic picture of the interactions between lipid storage, mobilization and signalling, which operate during normal cell function and are altered in many important diseases. The development of methods for imaging intracellular lipids can provide a dynamic real-time picture of how lipids are involved in complex signalling and other cell biology pathways; and how they ultimately regulate metabolic function/homoeostasis during early development. Some imaging modalities have the potential to be adapted for in vivo applications, and may enable the direct visualization of progression of pathogenesis of cardiometabolic disease after poor growth in early life.
Collapse
|
32
|
Peters J, Martinez N, Lehofer B, Prassl R. Low-density lipoproteins investigated under high hydrostatic pressure by elastic incoherent neutron scattering. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2017; 40:68. [PMID: 28733727 PMCID: PMC5589066 DOI: 10.1140/epje/i2017-11558-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/11/2017] [Indexed: 05/14/2023]
Abstract
Human low-density lipoprotein (LDL) is a highly complex nano-particle built up of various lipid classes and a single large protein moiety (apoB-100) owning essential physiological functions in the human body. Besides its vital role as a supplier of cholesterol and fat for peripheral tissues and cells, it is also a known key player in the formation of atherosclerosis. Due to these important roles in physiology and pathology the elucidation of structural and dynamical details is of great interest. In the current study we drew a broader picture of LDL dynamics using elastic incoherent neutron scattering (EINS) as a function of specified temperature and pressure points. We not only investigated a normolipidemic LDL sample, but also a triglyceride-rich and an oxidized one to mimic pathologic conditions as found under hyperlipidemic conditions or in atherosclerotic plaques, respectively. We could show that pressure has a significant effect on atomic motions in modified forms of LDL, whereas the normolipidemic sample seems to cope much better with high-pressure conditions irrespective of temperature. These findings might be explained by the altered lipid composition, which is either caused through elevated triglyceride content or modifications through lipid peroxidation.
Collapse
Affiliation(s)
- J Peters
- Univ. Grenoble Alpes, LiPhy, F-38044, Grenoble, France
- Institut Laue Langevin, F-38000, Grenoble, France
| | - N Martinez
- Institut Laue Langevin, F-38000, Grenoble, France
- Univ. Grenoble Alpes, IBS, F-38000, Grenoble, France
| | - B Lehofer
- Institute of Biophysics, Medical University of Graz, A-8010, Graz, Austria
| | - R Prassl
- Institute of Biophysics, Medical University of Graz, A-8010, Graz, Austria.
| |
Collapse
|
33
|
Maric S, Lind TK, Lyngsø J, Cárdenas M, Pedersen JS. Modeling Small-Angle X-ray Scattering Data for Low-Density Lipoproteins: Insights into the Fatty Core Packing and Phase Transition. ACS NANO 2017; 11:1080-1090. [PMID: 28048943 DOI: 10.1021/acsnano.6b08089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Atherosclerosis and its clinical consequences are the leading cause of death in the western hemisphere. While many studies throughout the last decades have aimed at understanding the disease, the clinical markers in use today still fail to accurately predict the risks. The role of the current main clinical indicator, low density lipoprotein (LDL), in depositing fat to the vessel wall is believed to be the onset of the process. However, many subfractions of the LDL, which differ both in structure and composition, are present in the blood and among different individuals. Understanding the relationship between LDL structure and composition is key to unravel the specific role of various LDL components in the development and/or prevention of atherosclerosis. Here, we describe a model for analyzing small-angle X-ray scattering data for rapid and robust structure determination for the LDL. The model not only gives the overall structure but also the particular internal layering of the fats inside the LDL core. Thus, the melting of the LDL can be followed in situ as a function of temperature for samples extracted from healthy human patients and purified using a double protocol based on ultracentrifugation and size-exclusion chromatography. The model provides information on: (i) the particle-specific melting temperature of the core lipids, (ii) the structural organization of the core fats inside the LDL, (iii) the overall shape of the particle, and (iv) the flexibility and overall conformation of the outer protein/hydrophilic layer at a given temperature as governed by the organization of the core. The advantage of this method over other techniques such as cryo-TEM is the possibility of in situ experiments under near-physiological conditions which can be performed relatively fast (minutes at home source, seconds at synchrotron). This approach now allows the monitoring of structural changes in the LDL upon different stresses from the environment, such as changes in temperature, oxidation, or external agents used or currently in development against atherosclerotic plaque build-up and which are targeting the LDL.
Collapse
Affiliation(s)
- Selma Maric
- Biofilms- Research Center for Biointerfaces, Dept. of Biomedical Science, Faculty of Health and Society, Malmö University , Malmö 20506, Sweden
| | - Tania Kjellerup Lind
- Biofilms- Research Center for Biointerfaces, Dept. of Biomedical Science, Faculty of Health and Society, Malmö University , Malmö 20506, Sweden
| | - Jeppe Lyngsø
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University , 8000 Aarhus, Denmark
| | - Marité Cárdenas
- Biofilms- Research Center for Biointerfaces, Dept. of Biomedical Science, Faculty of Health and Society, Malmö University , Malmö 20506, Sweden
| | - Jan Skov Pedersen
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University , 8000 Aarhus, Denmark
| |
Collapse
|
34
|
Yu Y, Kuang YL, Lei D, Zhai X, Zhang M, Krauss RM, Ren G. Polyhedral 3D structure of human plasma very low density lipoproteins by individual particle cryo-electron tomography1. J Lipid Res 2016; 57:1879-1888. [PMID: 27538822 PMCID: PMC5036368 DOI: 10.1194/jlr.m070375] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Indexed: 12/21/2022] Open
Abstract
Human VLDLs assembled in the liver and secreted into the circulation supply energy to peripheral tissues. VLDL lipolysis yields atherogenic LDLs and VLDL remnants that strongly correlate with CVD. Although the composition of VLDL particles has been well-characterized, their 3D structure is elusive because of their variations in size, heterogeneity in composition, structural flexibility, and mobility in solution. Here, we employed cryo-electron microscopy and individual-particle electron tomography to study the 3D structure of individual VLDL particles (without averaging) at both below and above their lipid phase transition temperatures. The 3D reconstructions of VLDL and VLDL bound to antibodies revealed an unexpected polyhedral shape, in contrast to the generally accepted model of a spherical emulsion-like particle. The smaller curvature of surface lipids compared with HDL may also reduce surface hydrophobicity, resulting in lower binding affinity to the hydrophobic distal end of the N-terminal β-barrel domain of cholesteryl ester transfer protein (CETP) compared with HDL. The directional binding of CETP to HDL and VLDL may explain the function of CETP in transferring TGs and cholesteryl esters between these particles. This first visualization of the 3D structure of VLDL could improve our understanding of the role of VLDL in atherogenesis.
Collapse
Affiliation(s)
- Yadong Yu
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Yu-Lin Kuang
- Atherosclerosis Research, Children's Hospital Oakland Research Institute, Oakland, CA 94609
| | - Dongsheng Lei
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Xiaobo Zhai
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Meng Zhang
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Ronald M Krauss
- Atherosclerosis Research, Children's Hospital Oakland Research Institute, Oakland, CA 94609
| | - Gang Ren
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720.
| |
Collapse
|
35
|
Balatskaya MN, Balatskii AV, Sharonov GV, Tkachuk VA. T-cadherin as a novel receptor regulating metabolism in the blood vessel and heart cells: from structure to function. J EVOL BIOCHEM PHYS+ 2016. [DOI: 10.1134/s0022093016020010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
Glukhova OE, Prytkova TR, Savostyanov GV. Simulation of High Density Lipoprotein Behavior on a Few Layer Graphene Undergoing Non-Uniform Mechanical Load. J Phys Chem B 2016; 120:3593-600. [DOI: 10.1021/acs.jpcb.5b12648] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Olga E. Glukhova
- Saratov State University, Astrakhanskaya 83, 410012, Saratov, Russia
| | - Tatiana R. Prytkova
- Schmid College of Science & Technology, Chapman University, Orange, California 92866, United States
| | | |
Collapse
|
37
|
Pan L, Segrest JP. Computational studies of plasma lipoprotein lipids. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2401-2420. [PMID: 26969087 DOI: 10.1016/j.bbamem.2016.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 12/27/2022]
Abstract
Plasma lipoproteins are macromolecular assemblies of proteins and lipids found in the blood. The lipid components of lipoproteins are amphipathic lipids such as phospholipids (PLs), and unesterified cholesterols (UCs) and hydrophobic lipids such as cholesteryl esters (CEs) and triglycerides (TGs). Since lipoproteins are soft matter supramolecular assemblies easily deformable by thermal fluctuations and they also exist in varying densities and protein/lipid components, a detailed understanding of their structure/function is experimentally difficult. Molecular dynamics (MD) simulation has emerged as a particularly promising way to explore the structure and dynamics of lipoproteins. The purpose of this review is to survey the current status of computational studies of the lipid components of the lipoproteins. Computational studies aim to explore three levels of complexity for the 3-dimensional structural dynamics of lipoproteins at various metabolic stages: (i) lipoprotein particles consist of protein with minimal lipid; (ii) lipoprotein particles consist of PL-rich discoidal bilayer-like lipid particles; (iii) mature circulating lipoprotein particles consist of CE-rich or TG-rich spheroidal lipid-droplet-like particles. Due to energy barriers involved in conversion between these species, other biomolecules also participate in lipoprotein biological assembly. For example: (i) lipid-poor apolipoprotein A-I (apoA-I) interacts with ATP-binding cassette transporter A1 (ABCA1) to produce nascent discoidal high density lipoprotein (dHDL) particles; (ii) lecithin-cholesterol acyltransferase (LCAT) mediates the conversion of UC to CE in dHDL, driving spheroidal HDL (sHDL) formation; (iii) transfer proteins, cholesterol ester transfer protein (CETP) and phospholipid transfer protein (PLTP), transfer both CE and TG and PL, respectively, between lipoprotein particles. Computational studies have the potential to explore different lipoprotein particles at each metabolic stage in atomistic detail. This review discusses the current status of computational methods including all-atom MD (AAMD), coarse-grain MD (CGMD), and MD-simulated annealing (MDSA) and their applications in lipoprotein structural dynamics and biological assemblies. Results from MD simulations are discussed and compared across studies in order to identify key findings, controversies, issues and future directions. This article is part of a Special Issue entitled: Biosimulations edited by Ilpo Vattulainen and Tomasz Róg.
Collapse
Affiliation(s)
- Lurong Pan
- Division of Gerontology, Geriatrics, & Palliative Care, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jere P Segrest
- Division of Gerontology, Geriatrics, & Palliative Care, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
38
|
Cherkouk C, Rebohle L, Lenk J, Keller A, Ou X, Laube M, Neuber C, Haase-Kohn C, Skorupa W, Pietzsch J. Controlled immobilization of His-tagged proteins for protein-ligand interaction experiments using Ni2+-NTA layer on glass surfaces. Clin Hemorheol Microcirc 2016; 61:523-39. [DOI: 10.3233/ch-151950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Charaf Cherkouk
- Technische Universität Bergakademie Freiberg, Institute of Experimental Physics, Freiberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Ion Beam Physics and Materials Research, Dresden, Germany
| | - Lars Rebohle
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Ion Beam Physics and Materials Research, Dresden, Germany
| | - Jens Lenk
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Adrian Keller
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Ion Beam Physics and Materials Research, Dresden, Germany
| | - Xin Ou
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Ion Beam Physics and Materials Research, Dresden, Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Cathleen Haase-Kohn
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Wolfgang Skorupa
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Ion Beam Physics and Materials Research, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
39
|
van Niel G, Bergam P, Di Cicco A, Hurbain I, Lo Cicero A, Dingli F, Palmulli R, Fort C, Potier MC, Schurgers LJ, Loew D, Levy D, Raposo G. Apolipoprotein E Regulates Amyloid Formation within Endosomes of Pigment Cells. Cell Rep 2015; 13:43-51. [PMID: 26387950 DOI: 10.1016/j.celrep.2015.08.057] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 07/02/2015] [Accepted: 08/20/2015] [Indexed: 01/20/2023] Open
Abstract
Accumulation of toxic amyloid oligomers is a key feature in the pathogenesis of amyloid-related diseases. Formation of mature amyloid fibrils is one defense mechanism to neutralize toxic prefibrillar oligomers. This mechanism is notably influenced by apolipoprotein E variants. Cells that produce mature amyloid fibrils to serve physiological functions must exploit specific mechanisms to avoid potential accumulation of toxic species. Pigment cells have tuned their endosomes to maximize the formation of functional amyloid from the protein PMEL. Here, we show that ApoE is associated with intraluminal vesicles (ILV) within endosomes and remain associated with ILVs when they are secreted as exosomes. ApoE functions in the ESCRT-independent sorting mechanism of PMEL onto ILVs and regulates the endosomal formation of PMEL amyloid fibrils in vitro and in vivo. This process secures the physiological formation of amyloid fibrils by exploiting ILVs as amyloid nucleating platforms.
Collapse
Affiliation(s)
- Guillaume van Niel
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France; Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris 75248, France.
| | - Ptissam Bergam
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France; Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris 75248, France
| | - Aurelie Di Cicco
- Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris 75248, France; Institut Curie, PSL Research University, UMR168, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR 168, Paris 75231, France
| | - Ilse Hurbain
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France; Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris 75248, France
| | - Alessandra Lo Cicero
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris 75248, France
| | - Roberta Palmulli
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France
| | - Cecile Fort
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France
| | - Marie Claude Potier
- Institut du Cerveau et de la Moelle, CNRS UMR7225, INSERM U1127, UPMC Hôpital de la Pitié-Salpêtrière, 47 Bd de l'Hôpital, Paris 75013, France
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Paris 75248, France
| | - Daniel Levy
- Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris 75248, France; Institut Curie, PSL Research University, UMR168, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR 168, Paris 75231, France
| | - Graça Raposo
- Institut Curie, PSL Research University, UMR144, Centre de Recherche, 26 rue d'ULM, Paris 75231, France; Centre National de la Recherche Scientifique, UMR144, Paris 75248, France; Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris 75248, France
| |
Collapse
|
40
|
Van der Horst DJ, Rodenburg KW. Lipoprotein assembly and function in an evolutionary perspective. Biomol Concepts 2015; 1:165-83. [PMID: 25961995 DOI: 10.1515/bmc.2010.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circulatory fat transport in animals relies on members of the large lipid transfer protein (LLTP) superfamily, including mammalian apolipoprotein B (apoB) and insect apolipophorin II/I (apoLp-II/I). ApoB and apoLp-II/I, constituting the structural (non-exchangeable) basis for the assembly of various lipoproteins, acquire lipids through microsomal triglyceride-transfer protein, another LLTP family member, and bind them by means of amphipathic α-helical and β-sheet structural motifs. Comparative research reveals that LLTPs evolved from the earliest animals and highlights the structural adaptations in these lipid-binding proteins. Thus, in contrast to apoB, apoLp-II/I is cleaved post-translationally by a furin, resulting in the appearance of two non-exchangeable apolipoproteins in the single circulatory lipoprotein in insects, high-density lipophorin (HDLp). The remarkable structural similarities between mammalian and insect lipoproteins notwithstanding important functional differences relate to the mechanism of lipid delivery. Whereas in mammals, partial delipidation of apoB-containing lipoproteins eventually results in endocytic uptake of their remnants, mediated by members of the low-density lipoprotein receptor (LDLR) family, and degradation in lysosomes, insect HDLp functions as a reusable lipid shuttle capable of alternate unloading and reloading of lipid. Also, during muscular efforts (flight activity), an HDLp-based lipoprotein shuttle provides for the transport of lipid for energy generation. Although a lipophorin receptor - a homolog of LDLR - was identified that mediates endocytic uptake of HDLp during specific developmental periods, the endocytosed lipoprotein appears to be recycled in a transferrin-like manner. These data highlight that the functional adaptations in the lipoprotein lipid carriers in mammals and insects also emerge with regard to the functioning of their cognate receptors.
Collapse
|
41
|
Analogs of LDL Receptor Ligand Motifs in Dengue Envelope and Capsid Proteins as Potential Codes for Cell Entry. ACTA ACUST UNITED AC 2015; 2015. [PMID: 27123468 DOI: 10.1155/2015/646303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It is established that cell entry of low density lipoprotein particles (LLPs) containing Apo B100 and Apo E is mediated by receptors and GAGs. Receptor ligand motifs, XBBBXXBX, XBBXBX, and ΨBΨXB, and mono- and bipartite NLS sequences are abundant in Apo E and Apo B100 as well as in envelope and capsid proteins of Dengue viruses 1-4 (DENV1-4). Synthetic, fluorescence-labeled peptides of sequences in DENV2 envelope protein, and DENV3 capsid that include these motifs were used to conduct a qualitative assessment of cell binding and entry capacity using HeLa cells. DENV2 envelope peptide, Dsp2EP, 0564Gly-Gly0595, was shown to bind and remain at the cell surface. In contrast, DENV3 capsid protein peptide, Dsp3CP, 0002Asn-Gln0028, readily enters HeLa cells and accumulates at discrete loci in the nucleus. FITC-labeled dengue synthetic peptides colocalize with Low Density Lipoprotein-CM-DiI and Apo E-CM-DiI to a degree that suggests that Dengue viruses may utilize cell entry pathways used by LLPs.
Collapse
|
42
|
Zhang M, Charles R, Tong H, Zhang L, Patel M, Wang F, Rames MJ, Ren A, Rye KA, Qiu X, Johns DG, Charles MA, Ren G. HDL surface lipids mediate CETP binding as revealed by electron microscopy and molecular dynamics simulation. Sci Rep 2015; 5:8741. [PMID: 25737239 PMCID: PMC4348656 DOI: 10.1038/srep08741] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/29/2015] [Indexed: 02/07/2023] Open
Abstract
Cholesteryl ester transfer protein (CETP) mediates the transfer of cholesterol esters (CE) from atheroprotective high-density lipoproteins (HDL) to atherogenic low-density lipoproteins (LDL). CETP inhibition has been regarded as a promising strategy for increasing HDL levels and subsequently reducing the risk of cardiovascular diseases (CVD). Although the crystal structure of CETP is known, little is known regarding how CETP binds to HDL. Here, we investigated how various HDL-like particles interact with CETP by electron microscopy and molecular dynamics simulations. Results showed that CETP binds to HDL via hydrophobic interactions rather than protein-protein interactions. The HDL surface lipid curvature generates a hydrophobic environment, leading to CETP hydrophobic distal end interaction. This interaction is independent of other HDL components, such as apolipoproteins, cholesteryl esters and triglycerides. Thus, disrupting these hydrophobic interactions could be a new therapeutic strategy for attenuating the interaction of CETP with HDL.
Collapse
Affiliation(s)
- Meng Zhang
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - River Charles
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Huimin Tong
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Lei Zhang
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Mili Patel
- Centre for Vascular Research, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
| | - Francis Wang
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Matthew J Rames
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Amy Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Kerry-Anne Rye
- Centre for Vascular Research, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
| | | | | | - M Arthur Charles
- School of Medicine, University of California, San Francisco, California 94115, USA
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| |
Collapse
|
43
|
Oliveira CLP, Santos PR, Monteiro AM, Figueiredo Neto AM. Effect of oxidation on the structure of human low- and high-density lipoproteins. Biophys J 2015; 106:2595-605. [PMID: 24940777 DOI: 10.1016/j.bpj.2014.04.049] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/20/2014] [Accepted: 04/30/2014] [Indexed: 10/25/2022] Open
Abstract
This work presents a controlled study of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) structural changes due to in vitro oxidation with copper ions. The changes were studied by small-angle x-ray scattering (SAXS) and dynamic light scattering (DLS) techniques in the case of LDL and by SAXS, DLS, and Z-scan (ZS) techniques in the case of HDL. SAXS data were analyzed with a to our knowledge new deconvolution method. This method provides the electron density profile of the samples directly from the intensity scattering of the monomers. Results show that LDL particles oxidized for 18 h show significant structural changes when compared to nonoxidized particles. Changes were observed in the electrical density profile, in size polydispersity, and in the degree of flexibility of the APO-B protein on the particle. HDL optical results obtained with the ZS technique showed a decrease of the amplitude of the nonlinear optical signal as a function of oxidation time. In contrast to LDL results reported in the literature, the HDL ZS signal does not lead to a complete loss of nonlinear optical signal after 18 h of copper oxidation. Also, the SAXS results did not indicate significant structural changes due to oxidation of HDL particles, and DLS results showed that a small number of oligomers formed in the sample oxidized for 18 h. All experimental results for the HDL samples indicate that this lipoprotein is more resistant to the oxidation process than are LDL particles.
Collapse
|
44
|
Affiliation(s)
- Vitaly V. Chaban
- MEMPHYS − Center for Biomembrane Physics, Syddansk Universitet, Odense M., 5230, Denmark
| | - Himanshu Khandelia
- MEMPHYS − Center for Biomembrane Physics, Syddansk Universitet, Odense M., 5230, Denmark
| |
Collapse
|
45
|
Lancaster JL, Antonijevic T, Starobin JM. Ordering and stability in lipid droplets with applications to low-density lipoproteins. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2014; 89:062708. [PMID: 25019815 DOI: 10.1103/physreve.89.062708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Indexed: 06/03/2023]
Abstract
In this article, we present a framework for investigating the order-disorder transition in lipid droplets using the standard Ising model. While a single lipid droplet is itself a complex system whose constituent cholesteryl esters each possesses many degrees of freedom, we present justification for using this effective approach to isolate the underlying physics. It is argued that the behavior of the esters confined within lipid droplets is significantly different from that of a bulk system of similar esters, which is adequately described by continuum mean-field theory in the thermodynamic limit. When the droplet's shell is modeled as an elastic membrane, a simple picture emerges for a transition between two ordered phases within the core which is tuned by the strength of interactions between the esters. Triglyceride concentration is proposed as a variable which strongly influences the strength of interactions between cholesteryl esters within droplets. The possible relevance of this mechanism to the well known atherogenic nature of small low-density lipoprotein particles is discussed in detail.
Collapse
Affiliation(s)
- Jarrett L Lancaster
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, 2907 E. Lee Street, Greensboro, North Carolina 27401, USA
| | - Todor Antonijevic
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, 2907 E. Lee Street, Greensboro, North Carolina 27401, USA
| | - Joseph M Starobin
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, 2907 E. Lee Street, Greensboro, North Carolina 27401, USA
| |
Collapse
|
46
|
Antonijevic T, Lancaster JL, Starobin JM. Modeling order-disorder transition in Low-Density Lipoprotein. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2014; 2014:5220-5223. [PMID: 25571170 DOI: 10.1109/embc.2014.6944802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Low Density Lipoproteins (LDL) undergo a reversible order-disorder thermal transition close to biological temperature due to cooperative melting of the cholesteryl esters (CE) in the core of the LDL particle. We have noticed that chain-chain interactions between CE molecules are responsible for the stability of the ordered smectic phase; thus, we formulated a simple "coarse-grained" two-state model to describe the melting process. In this model only nearest neighbor interactions are allowed. On the basis of these assumptions we performed Metropolis Monte Carlo (MC) simulation in order to obtain the heat capacity curve. The resulting profile reveals well-known features of the systems with a finite size.
Collapse
|
47
|
Garewal M, Zhang L, Ren G. Optimized negative-staining protocol for examining lipid-protein interactions by electron microscopy. Methods Mol Biol 2013; 974:111-8. [PMID: 23404274 PMCID: PMC10546916 DOI: 10.1007/978-1-62703-275-9_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
A large number of proteins are capable of inserting themselves into lipids, and interacting with membranes, such as transmembrane proteins and apolipoproteins. Protein-lipid interactions have been identified as one of the keys in understanding biological processes, while the structure of proteins at the lipid-binding stage can provide evidence to help identify their roles and critical functions. However, structure determination of proteins at the lipid-binding stage is rather difficult, because conformational and compositional heterogeneities of the protein-lipid complexes are major barriers to unravel their structures using traditional methods, such as X-ray crystallography. Electron microscopy (EM) is an alternative approach to determine protein structure and has demonstrated a capability in visualizing lipid-protein interactions directly. Among various EM techniques, negative-staining (NS) is an easy, rapid, qualitative approach that is a well-established technique, frequently used in research laboratories. Conventional NS protocols, unfortunately, often generate artifacts with lipid-related proteins, such as the rouleau formation of lipoproteins. To overcome this artifact formation, Ren and his colleagues recently developed an optimized NS protocol that was validated by comparing images of lipoproteins from cryo-electron microscopy (cryo-EM). The optimized NS protocol could produce "near native-state" particle images and high contrast images of the protein in its lipid-binding state that is favorable for three-dimensional (3D) reconstruction by single-particle analysis and individual-particle electron tomography (IPET), suggesting this optimized protocol can be used widely to examine the structure of proteins at lipid-binding stage.
Collapse
Affiliation(s)
- Mark Garewal
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | | |
Collapse
|
48
|
Khachfe HM, Atkinson D. Conformation and stability properties of B17: I. Analytical investigations using circular dichroism. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2012; 41:639-46. [PMID: 22828936 DOI: 10.1007/s00249-012-0836-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 04/24/2012] [Accepted: 06/25/2012] [Indexed: 11/29/2022]
Abstract
Structural characterization of B17, the 17% N-terminal domain of apo B, was carried out using circular dichroic (CD) spectroscopy, where secondary and tertiary structures were studied as a function of temperature and pH. Mild acidic conditions that correlate with histidine protonation invoked a change in the α-helix and random coil contents of the protein, with no apparent change in the β-sheet structural content. Specific changes in the structure of the protein that occur in response to temperature were also investigated to understand the stability and conformational changes of B17. Far- and near-UV CDs were used to probe the thermal changes in the protein. The protonation of some histidine residues was attributed to underlie the increase in the helical content of the protein.
Collapse
|
49
|
Hymel D, Peterson BR. Synthetic cell surface receptors for delivery of therapeutics and probes. Adv Drug Deliv Rev 2012; 64:797-810. [PMID: 22401875 PMCID: PMC3359398 DOI: 10.1016/j.addr.2012.02.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/18/2012] [Accepted: 02/20/2012] [Indexed: 11/17/2022]
Abstract
Receptor-mediated endocytosis is a highly efficient mechanism for cellular uptake of membrane-impermeant ligands. Cells use this process to acquire nutrients, initiate signal transduction, promote development, regulate neurotransmission, and maintain homeostasis. Natural receptors that participate in receptor-mediated endocytosis are structurally diverse, ranging from large transmembrane proteins to small glycolipids embedded in the outer leaflet of cellular plasma membranes. Despite their vast structural differences, these receptors share common features of binding to extracellular ligands, clustering in dynamic membrane regions that pinch off to yield intracellular vesicles, and accumulation of receptor-ligand complexes in membrane-sealed endosomes. Receptors typically dissociate from ligands in endosomes and cycle back to the cell surface, whereas internalized ligands are usually delivered into lysosomes, where they are degraded, but some can escape and penetrate into the cytosol. Here, we review efforts to develop synthetic cell surface receptors, defined as nonnatural compounds, exemplified by mimics of cholesterol, that insert into plasma membranes, bind extracellular ligands including therapeutics, probes, and endogenous proteins, and engage endocytic membrane trafficking pathways. By mimicking natural mechanisms of receptor-mediated endocytosis, synthetic cell surface receptors have the potential to function as prosthetic molecules capable of seamlessly augmenting the endocytic uptake machinery of living mammalian cells.
Collapse
Affiliation(s)
- David Hymel
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, United States
| | - Blake R. Peterson
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, United States
| |
Collapse
|
50
|
Electron microscopy of nanoemulsions: An essential tool for characterisation and stability assessment. Micron 2012; 43:85-103. [DOI: 10.1016/j.micron.2011.07.014] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/18/2011] [Accepted: 07/19/2011] [Indexed: 12/12/2022]
|