1
|
Iwaide S, Murakami T, Sedghi Masoud N, Kobayashi N, Fortin JS, Miyahara H, Higuchi K, Chambers JK. Classification of amyloidosis and protein misfolding disorders in animals 2024: A review on pathology and diagnosis. Vet Pathol 2024:3009858241283750. [PMID: 39389927 DOI: 10.1177/03009858241283750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Amyloidosis is a group of diseases in which proteins become amyloid, an insoluble fibrillar aggregate, resulting in organ dysfunction. Amyloid deposition has been reported in various animal species. To diagnose and understand the pathogenesis of amyloidosis, it is important to identify the amyloid precursor protein involved in each disease. Although 42 amyloid precursor proteins have been reported in humans, little is known about amyloidosis in animals, except for a few well-described amyloid proteins, including amyloid A (AA), amyloid light chain (AL), amyloid β (Aβ), and islet amyloid polypeptide-derived amyloid. Recently, several types of novel amyloidosis have been identified in animals using immunohistochemistry and mass spectrometry-based proteomic analysis. Certain species are predisposed to specific types of amyloidosis, suggesting a genetic background for its pathogenesis. Age-related amyloidosis has also emerged due to the increased longevity of captive animals. In addition, experimental studies have shown that some amyloids may be transmissible. Accurate diagnosis and understanding of animal amyloidosis are necessary for appropriate therapeutic intervention and comparative pathological studies. This review provides an updated classification of animal amyloidosis, including associated protein misfolding disorders of the central nervous system, and the current understanding of their pathogenesis. Pathologic features are presented together with state-of-the-art diagnostic methods that can be applied for routine diagnosis and identification of novel amyloid proteins in animals.
Collapse
Affiliation(s)
- Susumu Iwaide
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | - Tomoaki Murakami
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | | | | | | | | | - Keiichi Higuchi
- Shinshu University, Matsumoto, Japan
- Meio University, Nago, Japan
| | | |
Collapse
|
2
|
Gouveia M, Schmidt C, Basilio PG, Aveiro SS, Domingues P, Xia K, Colón W, Vitorino R, Ferreira R, Santos M, Vieira SI, Ribeiro F. Exercise training decreases the load and changes the content of circulating SDS-resistant protein aggregates in patients with heart failure with reduced ejection fraction. Mol Cell Biochem 2024; 479:2711-2722. [PMID: 37902886 PMCID: PMC11455743 DOI: 10.1007/s11010-023-04884-z] [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: 07/25/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Heart failure (HF) often disrupts the protein quality control (PQC) system leading to protein aggregate accumulation. Evidence from tissue biopsies showed that exercise restores PQC system in HF; however, little is known about its effects on plasma proteostasis. AIM To determine the effects of exercise training on the load and composition of plasma SDS-resistant protein aggregates (SRA) in patients with HF with reduced ejection fraction (HFrEF). METHODS Eighteen patients with HFrEF (age: 63.4 ± 6.5 years; LVEF: 33.4 ± 11.6%) participated in a 12-week combined (aerobic plus resistance) exercise program (60 min/session, twice per week). The load and content of circulating SRA were assessed using D2D SDS-PAGE and mass spectrometry. Cardiorespiratory fitness, quality of life, and circulating levels of high-sensitive C-reactive protein, N-terminal pro-B-type natriuretic peptide (NT-proBNP), haptoglobin and ficolin-3, were also evaluated at baseline and after the exercise program. RESULTS The exercise program decreased the plasma SRA load (% SRA/total protein: 38.0 ± 8.9 to 36.1 ± 9.7%, p = 0.018; % SRA/soluble fraction: 64.3 ± 27.1 to 59.8 ± 27.7%, p = 0.003). Plasma SRA of HFrEF patients comprised 31 proteins, with α-2-macroglobulin and haptoglobin as the most abundant ones. The exercise training significantly increased haptoglobin plasma levels (1.03 ± 0.40 to 1.11 ± 0.46, p = 0.031), while decreasing its abundance in SRA (1.83 ± 0.54 × 1011 to 1.51 ± 0.59 × 1011, p = 0.049). Cardiorespiratory fitness [16.4(5.9) to 19.0(5.2) ml/kg/min, p = 0.002], quality of life, and circulating NT-proBNP [720.0(850.0) to 587.0(847.3) pg/mL, p = 0.048] levels, also improved after the exercise program. CONCLUSION Exercise training reduced the plasma SRA load and enhanced PQC, potentially via haptoglobin-mediated action, while improving cardiorespiratory fitness and quality of life of patients with HFrEF.
Collapse
Affiliation(s)
- Marisol Gouveia
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal.
| | - Cristine Schmidt
- Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - Priscilla Gois Basilio
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - Susana S Aveiro
- Mass Spectrometry Centre, Department of Chemistry, LAQV REQUIMTE, University of Aveiro, Aveiro, Portugal
- GreenCoLab - Green Ocean Association, University of Algarve, Faro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, Department of Chemistry, LAQV REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, USA
- Centre for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Wilfredo Colón
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, USA
- Centre for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Rui Vitorino
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
- Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Ferreira
- Department of Chemistry, QOPNA & LAQV-REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Mário Santos
- Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
- Serviço de Cardiologia, Hospital Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, UMIB, University of Porto, Porto, Portugal
| | - Sandra I Vieira
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Fernando Ribeiro
- School of Health Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
3
|
Tasaki M, Lavatelli F, Obici L, Obayashi K, Miyamoto T, Merlini G, Palladini G, Ando Y, Ueda M. Age-related amyloidosis outside the brain: A state-of-the-art review. Ageing Res Rev 2021; 70:101388. [PMID: 34116224 DOI: 10.1016/j.arr.2021.101388] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/26/2021] [Accepted: 06/04/2021] [Indexed: 02/01/2023]
Abstract
Inside and outside the brain, accumulation of amyloid fibrils plays key roles in the pathogenesis of fatal age-related diseases such as Alzheimer's and Parkinson's diseases and wild-type transthyretin amyloidosis. Although the incidence of all amyloidoses increases with age, for some types of amyloidosis aging is known as the main direct risk factor, and these types are typically diseases of elderly people. More than 10 different precursor proteins are known to cause age-associated amyloidosis; these proteins include amyloid β protein, α-synuclein, transthyretin, islet amyloid polypeptide, atrial natriuretic factor, and the newly discovered epidermal growth factor-containing fibulin-like extracellular matrix protein 1. Except for intracerebral amyloidoses, most age-related amyloidoses have been little studied. Indeed, in view of the increasing life expectancy in our societies, understanding how aging is involved in the process of amyloid fibril accumulation and the effects of amyloid deposits on the aging body is extremely important. In this review, we summarize current knowledge about the nature of amyloid precursor proteins, the prevalence, clinical manifestations, and pathogenesis of amyloidosis, and recent advances in our understanding of age-related amyloidoses outside the brain.
Collapse
|
4
|
Gisonno RA, Masson T, Ramella NA, Barrera EE, Romanowski V, Tricerri MA. Evolutionary and structural constraints influencing apolipoprotein A-I amyloid behavior. Proteins 2021; 90:258-269. [PMID: 34414600 DOI: 10.1002/prot.26217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
Apolipoprotein A-I (apoA-I) has a key function in the reverse cholesterol transport. However, aggregation of apoA-I single point mutants can lead to hereditary amyloid pathology. Although several studies have tackled the biophysical and structural consequences introduced by these mutations, there is little information addressing the relationship between the evolutionary and structural features that contribute to the amyloid behavior of apoA-I. We combined evolutionary studies, in silico mutagenesis and molecular dynamics (MD) simulations to provide a comprehensive analysis of the conservation and pathogenic role of the aggregation-prone regions (APRs) present in apoA-I. Sequence analysis demonstrated that among the four amyloidogenic regions described for human apoA-I, only two (APR1 and APR4) are evolutionary conserved across different species of Sarcopterygii. Moreover, stability analysis carried out with the FoldX engine showed that APR1 contributes to the marginal stability of apoA-I. Structural properties of full-length apoA-I models suggest that aggregation is avoided by placing APRs into highly packed and rigid portions of its native fold. Compared to silent variants extracted from the gnomAD database, the thermodynamic and pathogenic impact of amyloid mutations showed evidence of a higher destabilizing effect. MD simulations of the amyloid variant G26R evidenced the partial unfolding of the alpha-helix bundle with the concomitant exposure of APR1 to the solvent, suggesting an insight into the early steps involved in its aggregation. Our findings highlight APR1 as a relevant component for apoA-I structural integrity and emphasize a destabilizing effect of amyloid variants that leads to the exposure of this region.
Collapse
Affiliation(s)
- Romina A Gisonno
- Facultad de Ciencias Médicas, Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP, CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Tomas Masson
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM, CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Nahuel A Ramella
- Facultad de Ciencias Médicas, Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP, CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Exequiel E Barrera
- Group of Biomolecular Simulations, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Víctor Romanowski
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM, CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - M Alejandra Tricerri
- Facultad de Ciencias Médicas, Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP, CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
5
|
Tasaki M, Okada M, Yanagisawa A, Nomura T, Matsushita H, Ueda A, Inoue Y, Masuda T, Misumi Y, Yamashita T, Nakamura T, Miyamoto T, Obayashi K, Ando Y, Ueda M. Apolipoprotein AI amyloid deposits in the ligamentum flavum in patients with lumbar spinal canal stenosis. Amyloid 2021; 28:107-112. [PMID: 33305623 DOI: 10.1080/13506129.2020.1858404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Amyloidosis is a protein-misfolding disease characterised by insoluble amyloid deposits in the extracellular space of various organs and tissues, such as the brain, heart, kidneys, and ligaments. We previously reported the frequent occurrence of amyloid deposits in the ligament flavum in the presence of lumbar spinal canal stenosis (LSCS), which is a common spinal disorder in older individuals. Our earlier clinicopathological studies revealed that amyloid deposits derived from transthyretin (TTR) were involved in the pathogenesis of LSCS. ATTR amyloid was the most common form in the ligamentum flavum, but amyloid deposits that were not identified still existed in more than 50% of patients with LSCS. In this study, we found apolipoprotein AI (AApoAI) amyloid deposits in the ligamentum flavum of patients with LSCS. The deposits occurred in 12% of patients with LSCS. Biochemical studies revealed that the amyloid deposits consisted mainly of full-length ApoAI. As a notable finding, the lumbar ligamentum flavum of patients who had LSCS with double-positive amyloid deposits-positive for both ATTR and AApoAI-was significantly thicker than that of patients who had LSCS with single-positive-that is, positive for either ATTR or AApoAI-amyloid deposits. We thus suggest that lumbar AApoAI amyloid formation may enhance the pathological changes of lumbar ATTR amyloidosis in patients with LSCS.
Collapse
Affiliation(s)
- Masayoshi Tasaki
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto, Japan.,Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masamitsu Okada
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Toshiya Nomura
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroaki Matsushita
- Department of Amyloidosis Research, Nagasaki International University, Sasebo, Japan
| | - Akihiko Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuteru Inoue
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Teruaki Masuda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yohei Misumi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taro Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takayuki Nakamura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Miyamoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Konen Obayashi
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Ando
- Department of Amyloidosis Research, Nagasaki International University, Sasebo, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
6
|
The concept of protein folding/unfolding and its impacts on human health. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021. [PMID: 34090616 DOI: 10.1016/bs.apcsb.2021.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Proteins have evolved in specific 3D structures and play different functions in cells and determine various reactions and pathways. The newly synthesized amino acid chains once depart ribosome must crumple into three-dimensional structures so can be biologically active. This process of protein that makes a functional molecule is called protein folding. The protein folding is both a biological and a physicochemical process that depends on the sequence of it. In fact, this process occurs more complicated and in some cases and in exposure to some molecules like glucose (glycation), mistaken folding leads to amyloid structures and fatal disorders called conformational diseases. Such conditions are detected by the quality control system of the cell and these abnormal proteins undergo renovation or degradation. This scenario takes place by the chaperones, chaperonins, and Ubiquitin-proteasome complex. Understanding of protein folding mechanisms from different views including experimental and computational approaches has revealed some intermediate ensembles such as molten globule and has been subjected to biophysical and molecular biology attempts to know more about prevalent conformational diseases.
Collapse
|
7
|
Townsend DJ, Middleton DA, Ashton L. Raman Spectroscopy with 2D Perturbation Correlation Moving Windows for the Characterization of Heparin-Amyloid Interactions. Anal Chem 2020; 92:13822-13828. [PMID: 32935978 DOI: 10.1021/acs.analchem.0c02390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It has been shown extensively that glycosaminoglycan (GAG)-protein interactions can induce, accelerate, and impede the clearance of amyloid fibrils associated with systemic and localized amyloidosis. Obtaining molecular details of these interactions is fundamental to our understanding of amyloid disease. Consequently, there is a need for analytical approaches that can identify protein conformational transitions and simultaneously characterize heparin interactions. By combining Raman spectroscopy with two-dimensional (2D) perturbation correlation moving window (2DPCMW) analysis, we have successfully identified changes in protein secondary structure during pH- and heparin-induced fibril formation of apolipoprotein A-I (apoA-I) associated with atherosclerosis. Furthermore, from the 2DPCMW, we have identified peak shifts and intensity variations in Raman peaks arising from different heparan sulfate moieties, indicating that protein-heparin interactions vary at different heparin concentrations. Raman spectroscopy thus reveals new mechanistic insights into the role of GAGs during amyloid fibril formation.
Collapse
Affiliation(s)
- David J Townsend
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - David A Middleton
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Lorna Ashton
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| |
Collapse
|
8
|
Ma'arfi F, Chandra S, Fatima JE, Khan MY, Mir SS, Yusuf MA. Probing the Structure-Function relationship and amyloidogenic propensities in natural variants of apolipoprotein A-I. Biochem Biophys Rep 2020; 24:100815. [PMID: 33024841 PMCID: PMC7527581 DOI: 10.1016/j.bbrep.2020.100815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 11/25/2022] Open
Abstract
Background Apolipoprotein A-I (apoA-I) protects against atherosclerosis and participates in the removal of excess cellular cholesterol from peripheral organs. Several naturally occurring apoA-I mutations are associated with familial systemic amyloidosis, with deposition of amyloid aggregates in peripheral organs, resulting in multiple organ failure. Systematic studies on naturally occurring variants are needed to delineate their roles and involvement in pathogenesis. Methods We performed a comparative structure–function analysis of five naturally occurring apoA-I variants and the wild-type protein. Circular dichroism, Fourier-transform infrared spectroscopy, thioflavin T and congo red fluorescence assays, thermal, chemical, and proteolytic stability assays, and 1,2-Dimyristoyl-sn-glycero-3-phosphocholine clearance analyses were used to assess the effects of mutations on the structure, function, stability, aggregation, and proteolytic susceptibility of the proteins to explore the mechanisms underlying amyloidosis and hypercholesterolemia. Results We observed structural changes in the mutants independent of fibril formation, suggesting the influence of the surrounding environment. The mutants were involved in aggregate formation to varying degree; L170P, R173P, and V156E showed an increased propensity to aggregate under different physiological conditions. β sheet formation indicates that L170P and R173P participate in amyloid formation. Compared to WT, V156E and L170P exhibited higher capacity for lipid clearance. Conclusions The selected point mutations, including those outside the hot spot regions of apoA-I structure, perturb the physiochemical and conformational behavior of the protein, influencing its function. General significance The study provides insights into the structure–function relationships of naturally occurring apoA-I variants outside the hot spot mutation sites. Several apoA-I mutants are associated with systemic amyloidosis. Structure–function analysis of five apoA-I variants and wild-type protein was done. Point mutations alter the physicochemical behavior and conformation of the variants.
Collapse
Affiliation(s)
- Farah Ma'arfi
- Department of Bioengineering, Integral University, Kursi Road, Dasauli, Lucknow, 226026, India
| | - Subhash Chandra
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Jamal e Fatima
- Department of Bioengineering, Integral University, Kursi Road, Dasauli, Lucknow, 226026, India
| | - Mohd Yasir Khan
- Department of Biosciences, Integral University, Kursi Road, Dasauli, Lucknow, 226026, India
| | - Snober S. Mir
- Department of Bioengineering, Integral University, Kursi Road, Dasauli, Lucknow, 226026, India
| | - Mohd Aslam Yusuf
- Department of Bioengineering, Integral University, Kursi Road, Dasauli, Lucknow, 226026, India
- Corresponding author. ;
| |
Collapse
|
9
|
Comparison of Patterns of Coronary Artery Disease in Patients With Heart Failure by Cardiac Amyloidosis Status. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2020; 27:31-35. [PMID: 33008788 DOI: 10.1016/j.carrev.2020.09.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND/PURPOSE The aim of this study is to characterize the pattern and the severity of coronary artery lesions in cardiac amyloidosis. METHODS We retrospectively compared patients with heart failure who tested positive (i.e., biopsy or gene tests - HF/CA+) against those who tested negative (HF/CA-) for cardiac amyloidosis. Groups were compared demographically and angiographically for qualitative and quantitative variables to determine patterns of involvement in the major epicardial coronary vessels. RESULTS The study included 110 heart failure patients, of whom 55 patients (88 lesions) were in the HF/CA+ group, and 55 patients (66 lesions) were HF/CA-. Despite the advanced age of HF/CA+ patients (74.5 ± 11.0 years vs. 54.1 ± 15.0 years; p = 0.05), no severe calcification was found in the HF/CA+ group (0.0% vs. 4.5%; p = 0.018). The HF/CA+ group also had fewer ostial lesions (3.4% vs. 15.1%; p = 0.0095) and a higher, albeit not significant, Thrombolysis in Myocardial Infarction frame count (30.4 ± 12.6 vs. 26.6 ± 11 frames; p = 0.06). In the HF/CA+ group, men had a significant number of tandem lesions compared to women (14.5% vs 0.0%, p = 0.02). CONCLUSIONS Overall, heart failure patients with cardiac amyloidosis were older but were found to have less calcified lesions, less ostial involvement, and a reduced anterograde coronary blood flow. This is the first report examining coronary lesions in heart failure patients with cardiac amyloidosis.
Collapse
|
10
|
Gisonno RA, Prieto ED, Gorgojo JP, Curto LM, Rodriguez ME, Rosú SA, Gaddi GM, Finarelli GS, Cortez MF, Schinella GR, Tricerri MA, Ramella NA. Fibrillar conformation of an apolipoprotein A-I variant involved in amyloidosis and atherosclerosis. Biochim Biophys Acta Gen Subj 2020; 1864:129515. [PMID: 31904503 DOI: 10.1016/j.bbagen.2020.129515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/21/2019] [Accepted: 12/30/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Different protein conformations may be involved in the development of clinical manifestations associated with human amyloidosis. Although a fibrillar conformation is usually the signature of damage in the tissues of patients, it is not clear whether this species is per se the cause or the consequence of the disease. Hereditary amyloidosis due to variants of apolipoprotein A-I (apoA-I) with a substitution of a single amino acid is characterized by the presence of fibrillar protein within the lesions. Thus mutations result in increased protein aggregation. Here we set up to characterize the folding of a natural variant with a mutation leading to a deletion at position 107 (apoA-I Lys107-0). Patients carrying this variant show amyloidosis and severe atherosclerosis. METHODS We oxidized this variant under controlled concentrations of hydrogen peroxide and analyzed the structure obtained after 30-day incubation by fluorescence, circular dichroism and microscopy approaches. Neutrophils activation was characterized by confocal microscopy. RESULTS We obtained a high yield of well-defined stable fibrillar structures of apoA-I Lys107-0. In an in vitro neutrophils system, we were able to detect the induction of Neutrophils Extracellular Traps (NETs) when we incubated with oxidized apoA-I variants. This effect was exacerbated by the fibrillar structure of oxidized Lys 107-0. CONCLUSIONS We conclude that a pro-inflammatory microenvironment could result in the formation of aggregation-prone species, which, in addition may induce a positive feed-back in the activation of an inflammatory response. GENERAL SIGNIFICANCE These events may explain a close association between amyloidosis due to apoA-I Lys107-0 and atherosclerosis.
Collapse
Affiliation(s)
- Romina A Gisonno
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | - Eduardo D Prieto
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), La Plata, Argentina
| | - Juan P Gorgojo
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), La Plata, Argentina
| | - Lucrecia M Curto
- Instituto de Química y Fisicoquímica Biológicas "Profesor Alejandro C. Paladini" (IQUIFIB) y Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CABA, Argentina
| | - M Eugenia Rodriguez
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), La Plata, Argentina
| | - Silvana A Rosú
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | - Gisela M Gaddi
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | | | - M Fernanda Cortez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina
| | - Guillermo R Schinella
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | - M Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina.
| | - Nahuel A Ramella
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina.
| |
Collapse
|
11
|
Howlett GJ, Ryan TM, Griffin MD. Lipid-apolipoprotein interactions in amyloid fibril formation and relevance to atherosclerosis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:502-507. [DOI: 10.1016/j.bbapap.2018.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/06/2018] [Accepted: 08/27/2018] [Indexed: 01/08/2023]
|
12
|
Hellberg S, Silvola JMU, Liljenbäck H, Kiugel M, Eskola O, Hakovirta H, Hörkkö S, Morisson-Iveson V, Hirani E, Saukko P, Ylä-Herttuala S, Knuuti J, Saraste A, Roivainen A. Amyloid-Targeting PET Tracer [ 18F]Flutemetamol Accumulates in Atherosclerotic Plaques. Molecules 2019; 24:molecules24061072. [PMID: 30893771 PMCID: PMC6471324 DOI: 10.3390/molecules24061072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/09/2019] [Accepted: 03/14/2019] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is characterized by the accumulation of oxidized lipids in the artery wall, which triggers an inflammatory response. Oxidized low-density lipoprotein (ox-LDL) presents amyloid-like structural properties, and different amyloid species have recently been recognized in atherosclerotic plaques. Therefore, we studied the uptake of the amyloid imaging agent [18F]Flutemetamol in atherosclerotic plaques. The binding of [18F]Flutemetamol to human carotid artery plaque was studied in vitro. In vivo uptake of the tracer was studied in hypercholesterolemic IGF-II/LDLR−/−ApoB100/100 mice and C57BL/6N controls. Tracer biodistribution was studied in vivo with PET/CT, and ex vivo by gamma counter and digital ex vivo autoradiography. The presence of amyloid, ox-LDL, and macrophages in the plaques was examined by immunohistochemistry. [18F]Flutemetamol showed specific accumulation in human carotid plaque, especially in areas positive for amyloid beta. The aortas of IGF-II/LDLR−/−ApoB100/100 mice showed large thioflavin-S-positive atherosclerotic plaques containing ox-LDL and macrophages. Autoradiography revealed 1.7-fold higher uptake in the plaques than in a lesion-free vessel wall, but no difference in aortic tissue uptake between mouse strains were observed in the in vivo PET/CT. In conclusion, [18F]Flutemetamol binds to amyloid-positive areas in human atherosclerotic plaques. Further studies are warranted to clarify the uptake mechanisms, and the potential of the tracer for in vivo imaging of atherosclerosis in patients.
Collapse
Affiliation(s)
- Sanna Hellberg
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
| | | | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, FI-20520 Turku, Finland.
| | - Max Kiugel
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
| | - Olli Eskola
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
| | - Harri Hakovirta
- Department of Vascular Surgery, Turku University Hospital, FI-20520 Turku, Finland.
| | - Sohvi Hörkkö
- Medical Research Center and Nordlab Oulu, University Hospital and Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland.
| | | | - Ella Hirani
- GE Healthcare Ltd., Chalfont St Giles HP8 4SP, UK.
| | - Pekka Saukko
- Department of Pathology and Forensic Medicine, University of Turku, FI-20520 Turku, Finland.
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70210 Kuopio, Finland.
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
| | - Antti Saraste
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland.
- Heart Center, Turku University Hospital, FI-20520 Turku, Finland.
- Department of Clinical Medicine, University of Turku, FI-20520 Turku, Finland.
| | - Anne Roivainen
- Turku PET Centre, University of Turku, FI-20520 Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, FI-20520 Turku, Finland.
| |
Collapse
|
13
|
Coffey AR, Kanke M, Smallwood TL, Albright J, Pitman W, Gharaibeh RZ, Hua K, Gertz E, Biddinger SB, Temel RE, Pomp D, Sethupathy P, Bennett BJ. microRNA-146a-5p association with the cardiometabolic disease risk factor TMAO. Physiol Genomics 2019; 51:59-71. [PMID: 30633643 DOI: 10.1152/physiolgenomics.00079.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Trimethylamine-N-oxide (TMAO), a microbial choline metabolism byproduct that is processed in the liver and excreted into circulation, is associated with increased atherosclerotic lesion formation and cardiovascular disease risk. Genetic regulators of TMAO levels are largely unknown. In the present study, we used 288 mice from a genetically heterogeneous mouse population [Diversity Outbred (DO)] to determine hepatic microRNA associations with TMAO in the context of an atherogenic diet. We also validated findings in two additional animal models of atherosclerosis: liver-specific insulin receptor knockout mice fed a chow diet (LIRKO) and African green monkeys fed high-fat/high-cholesterol diet. Small RNA-sequencing analysis in DO mice, LIRKO mice, and African green monkeys identified only one hepatic microRNA (miR-146a-5p) that is aberrantly expressed across all three models. Moreover, miR-146a-5p levels are associated with circulating TMAO after atherogenic diet in each of these models. We also performed high-resolution genetic mapping and identified a novel quantitative trait locus on Chromosome 12 for TMAO levels. This interval includes two genes, Numb and Dlst, which are inversely correlated with both miR-146a and TMAO and are predicted targets of miR-146a. Both of these genes have been validated as direct targets of miR-146a, though in other cellular contexts. This is the first report to our knowledge of a link between miR-146 and TMAO. Our findings suggest that miR-146-5p, as well as one or more genes at the Chromosome 12 QTL (possibly Numb or Dlst), is strongly linked to TMAO levels and likely involved in the control of atherosclerosis.
Collapse
Affiliation(s)
- Alisha R Coffey
- Curriculum in Genetics and Molecular Biology, University of North Carolina , Chapel Hill, North Carolina
| | - Matt Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University , Ithaca, New York
| | - Tangi L Smallwood
- Curriculum in Genetics and Molecular Biology, University of North Carolina , Chapel Hill, North Carolina
| | - Jody Albright
- Nutrition Research Institute, University of North Carolina, Kannapolis, North Carolina
| | - Wendy Pitman
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University , Ithaca, New York
| | - Raad Z Gharaibeh
- Department of Bioinformatics, University of North Carolina , Charlotte, North Carolina
| | - Kunjie Hua
- Department of Genetics, University of North Carolina , Chapel Hill, North Carolina
| | - Erik Gertz
- US Department of Agriculture, Agricultural Research Service Western Human Nutrition Research Center, Obesity and Metabolism Unit, Davis, California
| | - Sudha B Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Ryan E Temel
- Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky
| | - Daniel Pomp
- Department of Genetics, University of North Carolina , Chapel Hill, North Carolina
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University , Ithaca, New York
| | - Brian J Bennett
- US Department of Agriculture, Agricultural Research Service Western Human Nutrition Research Center, Obesity and Metabolism Unit, Davis, California
| |
Collapse
|
14
|
Witkowski A, Carta S, Lu R, Yokoyama S, Rubartelli A, Cavigiolio G. Oxidation of methionine residues in human apolipoprotein A-I generates a potent pro-inflammatory molecule. J Biol Chem 2019; 294:3634-3646. [PMID: 30635405 DOI: 10.1074/jbc.ra118.005663] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/27/2018] [Indexed: 02/06/2023] Open
Abstract
Amyloid deposits of apolipoprotein A-I (apoA-I) and inflammation are common in atherosclerotic arteries. In this study, we investigated the interplay between oxidation of apoA-I methionine residues (Met(O)-ApoA-I), a known amyloidogenic modification of apoA-I, and the inflammatory response of immune cells. Soluble pre-fibrillar Met(O)-ApoA-I, but not apoA-I, induced intracellular accumulation of pro-interleukin (IL)-1β and secretion of the pro-inflammatory cytokines tumor necrosis factor α (TNFα) and IL-6 in mouse bone marrow-derived macrophages (BMDMs) and human primary monocytes. Additionally, secretion of mature IL-1β was also activated in human monocytes. The pro-inflammatory activity of Met(O)-ApoA-I was Toll-like receptor 4 (TLR4)-dependent and CD36-independent and was solely determined by oxidation of apoA-I methionine residues, in particular Met-86 and Met-148. In contrast, amyloid fibrils or reconstituted high-density lipoproteins (HDLs) generated from Met(O)-ApoA-I did not induce cytokine production in BMDMs. Although lipid-free Met(O)-ApoA-I remained functional in extracting lipids from cells and generating HDL, it gained strong pro-inflammatory properties that may aggravate local inflammation in the arteries and atherosclerosis. Our study indicates that oxidation of apoA-I methionine residues produces a potent danger-associated molecular pattern capable of stimulating pro-inflammatory cytokine secretion at levels similar to those induced by known pathogen-associated molecular patterns, such as lipopolysaccharide.
Collapse
Affiliation(s)
- Andrzej Witkowski
- From the UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California 94609
| | - Sonia Carta
- the Cell Biology Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy, and
| | - Rui Lu
- Food and Nutritional Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Shinji Yokoyama
- Food and Nutritional Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Anna Rubartelli
- the Cell Biology Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy, and
| | - Giorgio Cavigiolio
- From the UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California 94609,
| |
Collapse
|
15
|
Townsend D, Hughes E, Akien G, Stewart KL, Radford SE, Rochester D, Middleton DA. Epigallocatechin-3-gallate remodels apolipoprotein A-I amyloid fibrils into soluble oligomers in the presence of heparin. J Biol Chem 2018; 293:12877-12893. [PMID: 29853648 PMCID: PMC6102129 DOI: 10.1074/jbc.ra118.002038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/25/2018] [Indexed: 11/06/2022] Open
Abstract
Amyloid deposits of WT apolipoprotein A-I (apoA-I), the main protein component of high-density lipoprotein, accumulate in atherosclerotic plaques where they may contribute to coronary artery disease by increasing plaque burden and instability. Using CD analysis, solid-state NMR spectroscopy, and transmission EM, we report here a surprising cooperative effect of heparin and the green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG), a known inhibitor and modulator of amyloid formation, on apoA-I fibrils. We found that heparin, a proxy for glycosaminoglycan (GAG) polysaccharides that co-localize ubiquitously with amyloid in vivo, accelerates the rate of apoA-I formation from monomeric protein and associates with insoluble fibrils. Mature, insoluble apoA-I fibrils bound EGCG (KD = 30 ± 3 μm; Bmax = 40 ± 3 μm), but EGCG did not alter the kinetics of apoA-I amyloid assembly from monomer in the presence or absence of heparin. EGCG selectively increased the mobility of specific backbone and side-chain sites of apoA-I fibrils formed in the absence of heparin, but the fibrils largely retained their original morphology and remained insoluble. By contrast, fibrils formed in the presence of heparin were mobilized extensively by the addition of equimolar EGCG, and the fibrils were remodeled into soluble 20-nm-diameter oligomers with a largely α-helical structure that were nontoxic to human umbilical artery endothelial cells. These results argue for a protective effect of EGCG on apoA-I amyloid associated with atherosclerosis and suggest that EGCG-induced remodeling of amyloid may be tightly regulated by GAGs and other amyloid co-factors in vivo, depending on EGCG bioavailability.
Collapse
Affiliation(s)
- David Townsend
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | - Eleri Hughes
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | - Geoffrey Akien
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | - Katie L Stewart
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - David Rochester
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | | |
Collapse
|
16
|
Witkowski A, Chan GKL, Boatz JC, Li NJ, Inoue AP, Wong JC, van der Wel PCA, Cavigiolio G. Methionine oxidized apolipoprotein A-I at the crossroads of HDL biogenesis and amyloid formation. FASEB J 2018; 32:3149-3165. [PMID: 29401604 DOI: 10.1096/fj.201701127r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Apolipoprotein A-I (apoA-I) shares with other exchangeable apolipoproteins a high level of structural plasticity. In the lipid-free state, the apolipoprotein amphipathic α-helices interact intra- and intermolecularly, providing structural stabilization by self-association. We have reported that lipid-free apoA-I becomes amyloidogenic upon physiologically relevant (myeloperoxidase-mediated) Met oxidation. In this study, we established that Met oxidation promotes amyloidogenesis by reducing the stability of apoA-I monomers and irreversibly disrupting self-association. The oxidized apoA-I monomers also exhibited increased cellular cholesterol release capacity and stronger association with macrophages, compared to nonoxidized apoA-I. Of physiologic relevance, preformed oxidized apoA-I amyloid fibrils induced amyloid formation in nonoxidized apoA-I. This process was enhanced when self-association of nonoxidized apoA-I was disrupted by thermal treatment. Solid state NMR analysis revealed that aggregates formed by seeded nonoxidized apoA-I were structurally similar to those formed by the oxidized protein, featuring a β-structure-rich amyloid fold alongside α-helices retained from the native state. In atherosclerotic lesions, the conditions that promote apoA-I amyloid formation are readily available: myeloperoxidase, active oxygen species, low pH, and high concentration of lipid-free apoA-I. Our results suggest that even partial Met oxidation of apoA-I can nucleate amyloidogenesis, thus sequestering and inactivating otherwise antiatherogenic and HDL-forming apoA-I.-Witkowski, A., Chan, G. K. L., Boatz, J. C., Li, N. J., Inoue, A. P., Wong, J. C., van der Wel, P. C. A., Cavigiolio, G. Methionine oxidized apolipoprotein A-I at the crossroads of HDL biogenesis and amyloid formation.
Collapse
Affiliation(s)
- Andrzej Witkowski
- Children's Hospital Oakland Research Institute (CHORI), Oakland, California, USA
| | - Gary K L Chan
- Children's Hospital Oakland Research Institute (CHORI), Oakland, California, USA
| | - Jennifer C Boatz
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nancy J Li
- Children's Hospital Oakland Research Institute (CHORI), Oakland, California, USA
| | - Ayuka P Inoue
- Children's Hospital Oakland Research Institute (CHORI), Oakland, California, USA
| | - Jaclyn C Wong
- Children's Hospital Oakland Research Institute (CHORI), Oakland, California, USA
| | | | - Giorgio Cavigiolio
- Children's Hospital Oakland Research Institute (CHORI), Oakland, California, USA
| |
Collapse
|
17
|
Molina-Sánchez P, Jorge I, Martinez-Pinna R, Blanco-Colio LM, Tarin C, Torres-Fonseca MM, Esteban M, Laustsen J, Ramos-Mozo P, Calvo E, Lopez JA, Ceniga MVD, Michel JB, Egido J, Andrés V, Vazquéz J, Meilhac O, Burillo E, Lindholt JS, Martin-Ventura JL. ApoA-I/HDL-C levels are inversely associated with abdominal aortic aneurysm progression. Thromb Haemost 2017; 113:1335-46. [DOI: 10.1160/th14-10-0874] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/21/2015] [Indexed: 12/18/2022]
Abstract
SummaryAbdominal aortic aneurysm (AAA) evolution is unpredictable, and there is no therapy except surgery for patients with an aortic size > 5 cm (large AAA). We aimed to identify new potential biomarkers that could facilitate prognosis and treatment of patients with AAA. A differential quantitative proteomic analysis of plasma proteins was performed in AAA patients at different stages of evolution [small AAA (aortic size=3�5cm) vs large AAA] using iTRAQ labelling, highthroughput nano-LC-MS/MS and a novel multi-layered statistical model. Among the proteins identified, ApoA-I was decreased in patients with large AAA compared to those with small AAA. These results were validated by ELISA on plasma samples from small (n=90) and large AAA (n=26) patients (150 ± 3 vs 133 ± 5 mg/dl, respectively, p< 0.001). ApoA-I levels strongly correlated with HDL-Cholesterol (HDL-C) concentration (r=0.9, p< 0.001) and showed a negative correlation with aortic size (r=-0.4, p< 0.01) and thrombus volume (r=-0.3, p< 0.01), which remained significant after adjusting for traditional risk factors. In a prospective study, HDL-C independently predicted aneurysmal growth rate in multiple linear regression analysis (n=122, p=0.008) and was inversely associated with need for surgical repair (Adjusted hazard ratio: 0.18, 95 % confidence interval: 0.04�0.74, p=0.018). In a nation-wide Danish registry, we found lower mean HDL-C concentration in large AAA patients (n=6,560) compared with patients with aorto-iliac occlusive disease (n=23,496) (0.89 ± 2.99 vs 1.59 ± 5.74 mmol/l, p< 0.001). Finally, reduced mean aortic AAA diameter was observed in AngII-infused mice treated with ApoA-I mimetic peptide compared with saline-injected controls. In conclusion, ApoAI/ HDL-C systemic levels are negatively associated with AAA evolution. Therapies targeting HDL functionality could halt AAA formation.
Collapse
|
18
|
Wang Y, Feng X, Shen B, Ma J, Zhao W. Is Vascular Amyloidosis Intertwined with Arterial Aging, Hypertension and Atherosclerosis? Front Genet 2017; 8:126. [PMID: 29085385 PMCID: PMC5649204 DOI: 10.3389/fgene.2017.00126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/04/2017] [Indexed: 12/21/2022] Open
Abstract
Vascular amyloidosis (VA) is a component of aging, but both VA and aging move forward together. Although, not all age-related molecules are involved with VA, some molecules are involved in a crosstalk between both of them. However, the cellular mechanism by which, vascular cells are phenotypically shifted to arterial remodeling, is not only involved in aging but also linked to VA. Additionally, patients with hypertension and atherosclerosis are susceptible to VA, while amyloidosis alone may provide fertile soil for the initiation and progression of subsequent hypertension and atherosclerosis. It is known that hypertension, atherosclerosis and amyloidosis can be viewed as accelerated aging. This review summarizes the available experimental and clinical evidence to help the reader to understand the advance and underlying mechanisms for VA involvement in and interaction with aging. Taken together, it is clear that VA, hypertension and atherosclerosis are closely intertwined with arterial aging as equal partners.
Collapse
Affiliation(s)
- Yushi Wang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaoxing Feng
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Botao Shen
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Jing Ma
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Waiou Zhao
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Lu C, Zuo K, Lu Y, Liang S, Huang X, Zeng C, Zhang J, An Y, Wang J. Apolipoprotein A-1-related amyloidosis 2 case reports and review of the literature. Medicine (Baltimore) 2017; 96:e8148. [PMID: 28953655 PMCID: PMC5626298 DOI: 10.1097/md.0000000000008148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
RATIONALE Apolipoprotein A-1 (ApoA-1)-related amyloidosis is characterized by the deposition of ApoA-1 in various organs and can be either hereditary or nonhereditary. It is rare and easily misdiagnosed. Renal involvement is common in hereditary ApoA-1 amyloidosis, but rare in the nonhereditary form. PATIENT CONCERNS We reported two cases with ApoA-1 amyloidosis, a 64-year-old man suffering from nephrotic syndrome and a 40-year-old man with nephrotic syndrome and splenomegaly. Renal biopsies revealed glomerular, interstitial and vascular amyloid deposits and positive phospholipase A2 receptor staining in the glomerular capillary loop in case 1, and mesangial amyloid deposits in case 2. DIAGNOSES After immunostaining failed to determine the specific amyloid protein, proteomic analysis of amyloid deposits by mass spectrometry was performed and demonstrated the ApoA-1 origin of the amyloid. Genetic testing revealed no mutation of the APOA1 gene in case 1 but a heterozygous mutation, Trp74Arg, in case 2. Case 1 was thus diagnosed as nonhereditary ApoA-1 associated renal amyloidosis with membranous nephropathy, and case 2 as hereditary ApoA-1 amyloidosis with multiorgan injuries (kidney and spleen) and a positive family history. INTERVENTIONS Case 1 was treated with glucocorticoid combined with cyclosporine. Case 2 was treated with calcitriol and angiotensin converting enzyme inhibitors. OUTCOMES Two cases were followed up for 5 months and 2 years, respectively; and case 1 was found to have attenuated proteinuria while case 2 had an elevation of cholestasis indices along with renal insufficiency. LESSONS Proteomic analysis by mass spectrometry of the amyloid deposits combined with genetic analysis can provide accurate diagnosis of ApoA-1 amyloidosis. Besides, these 2 cases expand our knowledge of ApoA-1-related renal amyloidosis.
Collapse
|
20
|
Townsend D, Hughes E, Hussain R, Siligardi G, Baldock S, Madine J, Middleton DA. Heparin and Methionine Oxidation Promote the Formation of Apolipoprotein A-I Amyloid Comprising α-Helical and β-Sheet Structures. Biochemistry 2017; 56:1632-1644. [DOI: 10.1021/acs.biochem.6b01120] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- David Townsend
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Eleri Hughes
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Rohanah Hussain
- Diamond Light Source Ltd., Diamond House, Harwell Science & Innovation Campus, Didcot OX11 0DE, Oxon, England
| | - Giuliano Siligardi
- Diamond Light Source Ltd., Diamond House, Harwell Science & Innovation Campus, Didcot OX11 0DE, Oxon, England
| | - Sarah Baldock
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Jillian Madine
- Department of Biochemistry, Institute
of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - David A. Middleton
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| |
Collapse
|
21
|
Iowa Mutant Apolipoprotein A-I (ApoA-IIowa) Fibrils Target Lysosomes. Sci Rep 2016; 6:30391. [PMID: 27464946 PMCID: PMC4964564 DOI: 10.1038/srep30391] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/30/2016] [Indexed: 01/01/2023] Open
Abstract
The single amino acid mutation G26R in human apolipoprotein A-I (apoA-IIowa) is the first mutation that was associated with familial AApoA1 amyloidosis. The N-terminal fragments (amino acid residues 1–83) of apoA-I containing this mutation deposit as amyloid fibrils in patients’ tissues and organs, but the mechanisms of cellular degradation and cytotoxicity have not yet been clarified. In this study, we demonstrated degradation of apoA-IIowa fibrils via the autophagy-lysosomal pathway in human embryonic kidney 293 cells. ApoA-IIowa fibrils induced an increase in lysosomal pH and the cytosolic release of the toxic lysosomal protease cathepsin B. The mitochondrial dysfunction caused by apoA-IIowa fibrils depended on cathepsin B and was ameliorated by increasing the degradation of apoA-IIowa fibrils. Thus, although apoA-IIowa fibril transport to lysosomes and fibril degradation in lysosomes may have occurred, the presence of an excess number of apoA-IIowa fibrils, more than the lysosomes could degrade, may be detrimental to cells. Our results thus provide evidence that the target of apoA-IIowa fibrils is lysosomes, and we thereby gained a novel insight into the mechanism of AApoA1 amyloidosis.
Collapse
|
22
|
Sakata N, Hoshii Y, Nakamura T, Kiyama M, Arai H, Omoto M, Morimatsu M, Ishihara T. Colocalization of Apolipoprotein AI in Various Kinds of Systemic Amyloidosis. J Histochem Cytochem 2016; 53:237-42. [PMID: 15684336 DOI: 10.1369/jhc.4a6387.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Apolipoprotein AI (apoAI), a major component of high-density lipoproteins, is one of the major amyloid fibril proteins and a minor constituent of the senile plaques observed in Alzheimer's disease. We examined colocalization of apoAI in various kinds of systemic amyloidosis in this study. Forty-three of 48 formalin-fixed paraffin-embedded heart specimens with various forms of systemic amyloidosis reacted immunohistochemically with anti-human apoAI antibody. ApoAI was also detected in water-extracted amyloid material by immunoblotting. In addition, we observed colocalization of apoAI and murine amyloid A (AA) amyloidosis in human apoAI transgenic mice. This is the first report of colocalization of apoAI with amyloid deposits in various forms of human systemic amyloidosis and murine AA amyloidosis in human apoAI transgenic mice. ApoAI may not always be a major component of amyloid fibrils, even when it is present in systemic amyloid deposits.
Collapse
Affiliation(s)
- Naohiro Sakata
- Department of Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Ryan TM, Griffin MDW, McGillivray DJ, Knott RB, Wood K, Masters CL, Kirby N, Curtain CC. Apolipoprotein C-II Adopts Distinct Structures in Complex with Micellar and Submicellar Forms of the Amyloid-Inhibiting Lipid-Mimetic Dodecylphosphocholine. Biophys J 2016; 110:85-94. [PMID: 26745412 PMCID: PMC4805880 DOI: 10.1016/j.bpj.2015.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/03/2015] [Accepted: 11/09/2015] [Indexed: 01/21/2023] Open
Abstract
The formation of amyloid deposits is a common feature of a broad range of diseases, including atherosclerosis, Alzheimer's disease, and Parkinson's disease. The basis and role of amyloid deposition in the pathogenesis of these diseases is still being defined, however an interesting feature of amyloidogenic proteins is that the majority of the pathologically associated proteins are involved in lipid homeostasis, be it in lipid transport, incorporation into membranes, or the regulation of lipid pathways. Thus, amyloid-forming proteins commonly bind lipids, and lipids are generally involved in the proper folding of these proteins. However, understanding of the basis for these lipid-related aspects of amyloidogenesis is lacking. Thus, we have used the apolipoprotein C-II amyloid model system in conjunction with x-ray and neutron scattering analyses to address this problem. Apolipoprotein C-II is a well-studied model system of systemic amyloid fibril formation, with a clear and well-defined pathway for fibril formation, where the effects of lipid interaction are characterized, particularly for the lipid mimetic dodecylphosphocholine. We show that the micellar state of an inhibitory lipid can have a very significant effect on protein conformation, with micelles stabilizing a particular α-helical structure, whereas submicellar lipids stabilize a very different dimeric, α-helical structure. These results indicate that lipids may have an important role in the development and progression of amyloid-related diseases.
Collapse
Affiliation(s)
- Timothy M Ryan
- Australian Synchrotron, Clayton, Victoria, Australia; The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia; The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington, New Zealand.
| | - Michael D W Griffin
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Duncan J McGillivray
- School of Chemical Science, The University of Auckland, Auckland, New Zealand; The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington, New Zealand
| | - Robert B Knott
- Australian Nuclear Science and Technology Organisation, New South Wales, Australia
| | - Kathleen Wood
- Australian Nuclear Science and Technology Organisation, New South Wales, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Nigel Kirby
- Australian Synchrotron, Clayton, Victoria, Australia
| | - Cyril C Curtain
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
24
|
Das M, Wilson CJ, Mei X, Wales TE, Engen JR, Gursky O. Structural Stability and Local Dynamics in Disease-Causing Mutants of Human Apolipoprotein A-I: What Makes the Protein Amyloidogenic? J Mol Biol 2015; 428:449-62. [PMID: 26562506 DOI: 10.1016/j.jmb.2015.10.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 01/27/2023]
Abstract
ApoA-I, the major protein of plasma high-density lipoprotein, removes cellular cholesterol and protects against atherosclerosis. ApoA-I mutations can cause familial amyloidosis, a life-threatening disease wherein N-terminal protein fragments form fibrils in vital organs. To unveil the protein misfolding mechanism and to understand why some mutations cause amyloidosis while others do not, we analyzed the structure, stability, and lipid-binding properties of naturally occurring mutants of full-length human apoA-I causing either amyloidosis (G26R, W50R, F71Y, and L170P) or aberrant lipid metabolism (L159R). Global and local protein conformation and dynamics in solution were assessed by circular dichroism, fluorescence, and hydrogen-deuterium exchange mass spectrometry. All mutants showed increased deuteration in residues 14-22, supporting our hypothesis that decreased protection of this major amyloid "hot spot" can trigger protein misfolding. In addition, L159R showed local helical unfolding near the mutation site, consistent with cleavage of this mutant in plasma to generate the labile 1-159 fragment. Together, the results suggest that reduced protection of the major amyloid "hot spot", combined with the structural integrity of the native helix bundle conformation, shifts the balance from protein clearance to β-aggregation. A delicate balance between the overall structural integrity of a globular protein and the local destabilization of its amyloidogenic segments may be a fundamental determinant of this and other amyloid diseases. Furthermore, mutation-induced conformational changes observed in the helix bundle, which comprises the N-terminal 75% of apoA-I, and its flexible C-terminal tail suggest the propagation of structural perturbations to distant sites via an unexpected template-induced ensemble-based mechanism, challenging the classical structure-based view.
Collapse
Affiliation(s)
- Madhurima Das
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA 02118, USA
| | - Christopher J Wilson
- Department of Chemistry & Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Xiaohu Mei
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA 02118, USA
| | - Thomas E Wales
- Department of Chemistry & Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - John R Engen
- Department of Chemistry & Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA 02118, USA.
| |
Collapse
|
25
|
Rosú SA, Rimoldi OJ, Prieto ED, Curto LM, Delfino JM, Ramella NA, Tricerri MA. Amyloidogenic propensity of a natural variant of human apolipoprotein A-I: stability and interaction with ligands. PLoS One 2015; 10:e0124946. [PMID: 25950566 PMCID: PMC4423886 DOI: 10.1371/journal.pone.0124946] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/19/2015] [Indexed: 12/18/2022] Open
Abstract
A number of naturally occurring mutations of human apolipoprotein A-I (apoA-I) have been associated with hereditary amyloidoses. The molecular mechanisms involved in amyloid-associated pathology remain largely unknown. Here we examined the effects of the Arg173Pro point mutation in apoA-I on the structure, stability, and aggregation propensity, as well as on the ability to bind to putative ligands. Our results indicate that the mutation induces a drastic loss of stability, and a lower efficiency to bind to phospholipid vesicles at physiological pH, which could determine the observed higher tendency to aggregate as pro-amyloidogenic complexes. Incubation under acidic conditions does not seem to induce significant desestabilization or aggregation tendency, neither does it contribute to the binding of the mutant to sodium dodecyl sulfate. While the binding to this detergent is higher for the mutant as compared to wt apoA-I, the interaction of the Arg173Pro variant with heparin depends on pH, being lower at pH 5.0 and higher than wt under physiological pH conditions. We suggest that binding to ligands as heparin or other glycosaminoglycans could be key events tuning the fine details of the interaction of apoA-I variants with the micro-environment, and probably eliciting the toxicity of these variants in hereditary amyloidoses.
Collapse
Affiliation(s)
- Silvana A. Rosú
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Omar J. Rimoldi
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Eduardo D. Prieto
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Universidad Nacional de La Plata-CONICET, La Plata, Buenos Aires, Argentina
| | - Lucrecia M. Curto
- Departamento de Química Biológica e Instituto de Bioquímica y Biofísica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - José M. Delfino
- Departamento de Química Biológica e Instituto de Bioquímica y Biofísica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nahuel A. Ramella
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - M. Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
26
|
Chan GKL, Witkowski A, Gantz DL, Zhang TO, Zanni MT, Jayaraman S, Cavigiolio G. Myeloperoxidase-mediated Methionine Oxidation Promotes an Amyloidogenic Outcome for Apolipoprotein A-I. J Biol Chem 2015; 290:10958-71. [PMID: 25759391 DOI: 10.1074/jbc.m114.630442] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Indexed: 11/06/2022] Open
Abstract
High plasma levels of apolipoprotein A-I (apoA-I) correlate with cardiovascular health, whereas dysfunctional apoA-I is a cause of atherosclerosis. In the atherosclerotic plaques, amyloid deposition increases with aging. Notably, apoA-I is the main component of these amyloids. Recent studies identified high levels of oxidized lipid-free apoA-I in atherosclerotic plaques. Likely, myeloperoxidase (MPO) secreted by activated macrophages in atherosclerotic lesions is the promoter of such apoA-I oxidation. We hypothesized that apoA-I oxidation by MPO levels similar to those present in the artery walls in atherosclerosis can promote apoA-I structural changes and amyloid fibril formation. ApoA-I was exposed to exhaustive chemical (H2O2) oxidation or physiological levels of enzymatic (MPO) oxidation and incubated at 37 °C and pH 6.0 to induce fibril formation. Both chemically and enzymatically oxidized apoA-I produced fibrillar amyloids after a few hours of incubation. The amyloid fibrils were composed of full-length apoA-I with differential oxidation of the three methionines. Met to Leu apoA-I variants were used to establish the predominant role of oxidation of Met-86 and Met-148 in the fibril formation process. Importantly, a small amount of preformed apoA-I fibrils was able to seed amyloid formation in oxidized apoA-I at pH 7.0. In contrast to hereditary amyloidosis, wherein specific mutations of apoA-I cause protein destabilization and amyloid deposition, oxidative conditions similar to those promoted by local inflammation in atherosclerosis are sufficient to transform full-length wild-type apoA-I into an amyloidogenic protein. Thus, MPO-mediated oxidation may be implicated in the mechanism that leads to amyloid deposition in the atherosclerotic plaques in vivo.
Collapse
Affiliation(s)
- Gary K L Chan
- From the Children's Hospital Oakland Research Institute, Oakland, California 94609
| | - Andrzej Witkowski
- From the Children's Hospital Oakland Research Institute, Oakland, California 94609
| | - Donald L Gantz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts 02118, and
| | - Tianqi O Zhang
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Martin T Zanni
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Shobini Jayaraman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts 02118, and
| | - Giorgio Cavigiolio
- From the Children's Hospital Oakland Research Institute, Oakland, California 94609,
| |
Collapse
|
27
|
Oskarsson ME, Paulsson JF, Schultz SW, Ingelsson M, Westermark P, Westermark GT. In vivo seeding and cross-seeding of localized amyloidosis: a molecular link between type 2 diabetes and Alzheimer disease. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:834-46. [PMID: 25700985 DOI: 10.1016/j.ajpath.2014.11.016] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/31/2014] [Accepted: 11/06/2014] [Indexed: 01/21/2023]
Abstract
Several proteins have been identified as amyloid forming in humans, and independent of protein origin, the fibrils are morphologically similar. Therefore, there is a potential for structures with amyloid seeding ability to induce both homologous and heterologous fibril growth; thus, molecular interaction can constitute a link between different amyloid forms. Intravenous injection with preformed fibrils from islet amyloid polypeptide (IAPP), proIAPP, or amyloid-beta (Aβ) into human IAPP transgenic mice triggered IAPP amyloid formation in pancreas in 5 of 7 mice in each group, demonstrating that IAPP amyloid could be enhanced through homologous and heterologous seeding with higher efficiency for the former mechanism. Proximity ligation assay was used for colocalization studies of IAPP and Aβ in islet amyloid in type 2 diabetic patients and Aβ deposits in brains of patients with Alzheimer disease. Aβ reactivity was not detected in islet amyloid although islet β cells express AβPP and convertases necessary for Aβ production. By contrast, IAPP and proIAPP were detected in cerebral and vascular Aβ deposits, and presence of proximity ligation signal at both locations showed that the peptides were <40 nm apart. It is not clear whether IAPP present in brain originates from pancreas or is locally produced. Heterologous seeding between IAPP and Aβ shown here may represent a molecular link between type 2 diabetes and Alzheimer disease.
Collapse
Affiliation(s)
- Marie E Oskarsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan F Paulsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Martin Ingelsson
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Per Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
28
|
Amyloid-Forming Properties of Human Apolipoproteins: Sequence Analyses and Structural Insights. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 855:175-211. [PMID: 26149931 DOI: 10.1007/978-3-319-17344-3_8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Apolipoproteins are protein constituents of lipoproteins that transport cholesterol and fat in circulation and are central to cardiovascular health and disease. Soluble apolipoproteins can transiently dissociate from the lipoprotein surface in a labile free form that can misfold, potentially leading to amyloid disease. Misfolding of apoA-I, apoA-II, and serum amyloid A (SAA) causes systemic amyloidoses, apoE4 is a critical risk factor in Alzheimer's disease, and apolipoprotein misfolding is also implicated in cardiovascular disease. To explain why apolipoproteins are over-represented in amyloidoses, it was proposed that the amphipathic α-helices, which form the lipid surface-binding motif in this protein family, have high amyloid-forming propensity. Here, we use 12 sequence-based bioinformatics approaches to assess amyloid-forming potential of human apolipoproteins and to identify segments that are likely to initiate β-aggregation. Mapping such segments on the available atomic structures of apolipoproteins helps explain why some of them readily form amyloid while others do not. Our analysis shows that nearly all amyloidogenic segments: (i) are largely hydrophobic, (ii) are located in the lipid-binding amphipathic α-helices in the native structures of soluble apolipoproteins, (iii) are predicted in both native α-helices and β-sheets in the insoluble apoB, and (iv) are predicted to form parallel in-register β-sheet in amyloid. Most of these predictions have been verified experimentally for apoC-II, apoA-I, apoA-II and SAA. Surprisingly, the rank order of the amino acid sequence propensity to form amyloid (apoB>apoA-II>apoC-II≥apoA-I, apoC-III, SAA, apoC-I>apoA-IV, apoA-V, apoE) does not correlate with the proteins' involvement in amyloidosis. Rather, it correlates directly with the strength of the protein-lipid association, which increases with increasing protein hydrophobicity. Therefore, the lipid surface-binding function and the amyloid-forming propensity are both rooted in apolipoproteins' hydrophobicity, suggesting that functional constraints make it difficult to completely eliminate pathogenic apolipoprotein misfolding. We propose that apolipoproteins have evolved protective mechanisms against misfolding, such as the sequestration of the amyloidogenic segments via the native protein-lipid and protein-protein interactions involving amphipathic α-helices and, in case of apoB, β-sheets.
Collapse
|
29
|
The Role of Lipid in Misfolding and Amyloid Fibril Formation by Apolipoprotein C-II. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 855:157-74. [DOI: 10.1007/978-3-319-17344-3_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
30
|
PARK SJ, KIM JY, TEOH CL, KANG NY, CHANG YT. New Targets of Molecular Imaging in Atherosclerosis: Prehension of Current Status. ANAL SCI 2015; 31:245-55. [DOI: 10.2116/analsci.31.245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Sung-Jin PARK
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Jun-Young KIM
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Chai Lean TEOH
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Nam-Young KANG
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Young-Tae CHANG
- Department of Chemistry & NUS MedChem Program of Life Sciences Institute, National University of Singapore
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| |
Collapse
|
31
|
Abstract
BACKGROUND Senile systemic amyloidosis (SSA) derived from wild-type transthyretin is a fairly common condition of old individuals, particularly men. The main presentation is by cardiac involvement, which can lead to severe restrictive cardiomyopathy. SSA is, however, a systemic disease, and amyloid deposits may appear in many other tissues but are thought to be without clinical symptoms outside the heart. Amyloid is a very common finding in cartilage and ligaments of elderly subjects, and transthyretin has been demonstrated in some deposits. Lumbar spinal stenosis is also a condition of usually elderly individuals in whom narrowing of the lumbar spinal canal leads to compression of nerves to the lower limbs. RESULTS We questioned whether lumbar spinal stenosis sometimes could be a manifestation of undiagnosed SSA. In this first report we have studied the presence of amyloid in material obtained at surgery for spinal stenosis in 26 patients. Amyloid was found in 25 subjects. Transthyretin was demonstrated immunohistochemically in 5 out of 15 studied resected tissues. Four of the positive materials were analyzed with Western blot revealing both full-length transthyretin (TTR) and C-terminal TTR fragments, typically seen in SSA. CONCLUSION We conclude that lumbar spinal stenosis quite frequently may be a consequence of SSA and that further studies are warranted.
Collapse
Affiliation(s)
- Per Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Ole B. Suhr
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Svante Berg
- Stockholm Spine Centre, Löwenströmska Hospital, Upplands Väsby, Sweden
| |
Collapse
|
32
|
Das M, Mei X, Jayaraman S, Atkinson D, Gursky O. Amyloidogenic mutations in human apolipoprotein A-I are not necessarily destabilizing - a common mechanism of apolipoprotein A-I misfolding in familial amyloidosis and atherosclerosis. FEBS J 2014; 281:2525-42. [PMID: 24702826 DOI: 10.1111/febs.12809] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/19/2014] [Accepted: 04/04/2014] [Indexed: 12/18/2022]
Abstract
High-density lipoproteins and their major protein, apolipoprotein A-I (apoA-I), remove excess cellular cholesterol and protect against atherosclerosis. However, in acquired amyloidosis, nonvariant full-length apoA-I deposits as fibrils in atherosclerotic plaques; in familial amyloidosis, N-terminal fragments of variant apoA-I deposit in vital organs, damaging them. Recently, we used the crystal structure of Δ(185-243)apoA-I to show that amyloidogenic mutations destabilize apoA-I and increase solvent exposure of the extended strand 44-55 that initiates β-aggregation. In the present study, we test this hypothesis by exploring naturally occurring human amyloidogenic mutations, W50R and G26R, within or close to this strand. The mutations caused small changes in the protein's α-helical content, stability, proteolytic pattern and protein-lipid interactions. These changes alone were unlikely to account for amyloidosis, suggesting the importance of other factors. Sequence analysis predicted several amyloid-prone segments that can initiate apoA-I misfolding. Aggregation studies using N-terminal fragments verified this prediction experimentally. Three predicted N-terminal amyloid-prone segments, mapped on the crystal structure, formed an α-helical cluster. Structural analysis indicates that amyloidogenic mutations or Met86 oxidation perturb native packing in this cluster. Taken together, the results suggest that structural perturbations in the amyloid-prone segments trigger α-helix to β-sheet conversion in the N-terminal ~ 75 residues forming the amyloid core. Polypeptide outside this core can be proteolysed to form 9-11 kDa N-terminal fragments found in familial amyloidosis. Our results imply that apoA-I misfolding in familial and acquired amyloidosis follows a similar mechanism that does not require significant structural destabilization or proteolysis. This novel mechanism suggests potential therapeutic interventions for apoA-I amyloidosis.
Collapse
Affiliation(s)
- Madhurima Das
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | |
Collapse
|
33
|
Castellano J, Badimon L, Llorente-Cortés V. Amyloid-β increases metallo- and cysteine protease activities in human macrophages. J Vasc Res 2013; 51:58-67. [PMID: 24335416 DOI: 10.1159/000356334] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/29/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND/AIMS Amyloid-β (Aβ) plays a crucial role in the onset and progression of atherosclerosis. Macrophages are a source of matrix metalloproteinases (MMPs), cysteine proteases and transforming growth factor (TGF)-β1 in the vascular wall. The aims of this study were to analyze the capacity of Aβ peptide (1-40) (Aβ40), Aβ peptide (1-42) (Aβ42) and fibrillar Aβ42 (fAβ42) to modulate the expression and activity of MMP-9, MMP-2 and tissue inhibitor of MMP-1 (TIMP-1) in human monocyte-derived macrophages (HMDM). Additionally, we analyzed whether Aβ internalization alters the secretion of cathepsin S (CatS) and TGF-β1 by macrophages. METHODS HMDM were exposed to native and fibrillar Aβ. MMPs and TIMP-1 expression was analyzed by real-time PCR, and MMP abundance by zymography. Protein levels of precursor and active forms of CatS were analyzed by Western blot and TGF-β1 levels by ELISA. RESULTS Aβ40, Aβ42 and especially fAβ42 strongly induced MMP-9/MMP-2 levels. Moreover, we showed enhanced active CatS and reduced TGF-β1 protein levels in the secretome of Aβ42 and fAβ42-exposed macrophages. CONCLUSIONS Aβ can regulate the proinflammatory state of human macrophages by inducing metallo- and cysteine protease levels and by reducing TGF-β1 secretion. These effects may be crucial in atherosclerosis progression.
Collapse
Affiliation(s)
- José Castellano
- Cardiovascular Research Center CSIC-ICCC, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | |
Collapse
|
34
|
Apolipoprotein A-I Helsinki promotes intracellular acyl-CoA cholesterol acyltransferase (ACAT) protein accumulation. Mol Cell Biochem 2013; 377:197-205. [DOI: 10.1007/s11010-013-1585-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/30/2013] [Indexed: 10/27/2022]
|
35
|
Burke KA, Yates EA, Legleiter J. Biophysical insights into how surfaces, including lipid membranes, modulate protein aggregation related to neurodegeneration. Front Neurol 2013; 4:17. [PMID: 23459674 PMCID: PMC3585431 DOI: 10.3389/fneur.2013.00017] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/09/2013] [Indexed: 11/13/2022] Open
Abstract
There are a vast number of neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD), associated with the rearrangement of specific proteins to non-native conformations that promotes aggregation and deposition within tissues and/or cellular compartments. These diseases are commonly classified as protein-misfolding or amyloid diseases. The interaction of these proteins with liquid/surface interfaces is a fundamental phenomenon with potential implications for protein-misfolding diseases. Kinetic and thermodynamic studies indicate that significant conformational changes can be induced in proteins encountering surfaces, which can play a critical role in nucleating aggregate formation or stabilizing specific aggregation states. Surfaces of particular interest in neurodegenerative diseases are cellular and subcellular membranes that are predominately comprised of lipid components. The two-dimensional liquid environments provided by lipid bilayers can profoundly alter protein structure and dynamics by both specific and non-specific interactions. Importantly for misfolding diseases, these bilayer properties can not only modulate protein conformation, but also exert influence on aggregation state. A detailed understanding of the influence of (sub)cellular surfaces in driving protein aggregation and/or stabilizing specific aggregate forms could provide new insights into toxic mechanisms associated with these diseases. Here, we review the influence of surfaces in driving and stabilizing protein aggregation with a specific emphasis on lipid membranes.
Collapse
Affiliation(s)
- Kathleen A Burke
- C. Eugene Bennett Department of Chemistry, West Virginia University Morgantown, WV, USA
| | | | | |
Collapse
|
36
|
Human apolipoprotein A-I natural variants: molecular mechanisms underlying amyloidogenic propensity. PLoS One 2012; 7:e43755. [PMID: 22952757 PMCID: PMC3429494 DOI: 10.1371/journal.pone.0043755] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/25/2012] [Indexed: 01/11/2023] Open
Abstract
Human apolipoprotein A-I (apoA-I)-derived amyloidosis can present with either wild-type (Wt) protein deposits in atherosclerotic plaques or as a hereditary form in which apoA-I variants deposit causing multiple organ failure. More than 15 single amino acid replacement amyloidogenic apoA-I variants have been described, but the molecular mechanisms involved in amyloid-associated pathology remain largely unknown. Here, we have investigated by fluorescence and biochemical approaches the stabilities and propensities to aggregate of two disease-associated apoA-I variants, apoA-IGly26Arg, associated with polyneuropathy and kidney dysfunction, and apoA-ILys107-0, implicated in amyloidosis in severe atherosclerosis. Results showed that both variants share common structural properties including decreased stability compared to Wt apoA-I and a more flexible structure that gives rise to formation of partially folded states. Interestingly, however, distinct features appear to determine their pathogenic mechanisms. ApoA-ILys107-0 has an increased propensity to aggregate at physiological pH and in a pro-inflammatory microenvironment than Wt apoA-I, whereas apoA-IGly26Arg elicited macrophage activation, thus stimulating local chronic inflammation. Our results strongly suggest that some natural mutations in apoA-I variants elicit protein tendency to aggregate, but in addition the specific interaction of different variants with macrophages may contribute to cellular stress and toxicity in hereditary amyloidosis.
Collapse
|
37
|
Ryan TM, Griffin MDW, Bailey MF, Schuck P, Howlett GJ. NBD-labeled phospholipid accelerates apolipoprotein C-II amyloid fibril formation but is not incorporated into mature fibrils. Biochemistry 2011; 50:9579-86. [PMID: 21985034 DOI: 10.1021/bi201192r] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human apolipoprotein (apo) C-II is one of several lipid-binding proteins that self-assemble into fibrils and accumulate in disease-related amyloid deposits. A general characteristic of these amyloid deposits is the presence of lipids, known to modulate individual steps in amyloid fibril formation. ApoC-II fibril formation is activated by submicellar phospholipids but inhibited by micellar lipids. We examined the mechanism for the activation by submicellar lipids using the fluorescently labeled, short-chain phospholipid 1-dodecyl-[(7-nitro-2-1,3-benzoxadiazol-4-yl)amino]-2-hydroxyglycero-3-phosphocholine (NBD-lyso-12-PC). Addition of submicellar NBD-lyso-12-PC increased the rate of fibril formation by apoC-II approximately 2-fold. Stopped flow kinetic analysis using fluorescence detection and low, non-fibril-forming concentrations of apoC-II indicated NBD-lyso-12-PC binds rapidly, on the millisecond time scale, followed by the slower formation of discrete apoC-II tetramers. Sedimentation velocity analysis showed NBD-lyso-12-PC binds to both apoC-II monomers and tetramers at approximately five sites per monomer with an average dissociation constant of approximately 10 μM. Mature apoC-II fibrils formed in the presence of NBD-lyso-12-PC were devoid of lipid, indicating a purely catalytic role for submicellar lipids in the activation of apoC-II fibril formation. These studies demonstrate the catalytic potential of small amphiphilic molecules in controlling protein folding and fibril assembly pathways.
Collapse
Affiliation(s)
- Timothy M Ryan
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
38
|
Jayaraman S, Abe-Dohmae S, Yokoyama S, Cavigiolio G. Impact of self-association on function of apolipoprotein A-I. J Biol Chem 2011; 286:35610-35623. [PMID: 21835924 DOI: 10.1074/jbc.m111.262485] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Self-association is an inherent property of the lipid-free forms of several exchangeable apolipoproteins, including apolipoprotein A-I (apoA-I), the main protein component of high density lipoproteins (HDL) and an established antiatherogenic factor. Monomeric lipid-free apoA-I is believed to be the biologically active species, but abnormal conditions, such as specific natural mutations or oxidation, produce an altered state of self-association that may contribute to apoA-I dysfunction. Replacement of the tryptophans of apoA-I with phenylalanines (ΔW-apoA-I) leads to unusually large and stable self-associated species. We took advantage of this unique solution property of ΔW-apoA-I to analyze the role of self-association in determining the structure and lipid-binding properties of apoA-I as well as ATP-binding cassette A1 (ABCA1)-mediated cellular lipid release, a relevant pathway in atherosclerosis. Monomeric ΔW-apoA-I and wild-type apoA-I activated ABCA1-mediated cellular lipid release with similar efficiencies, whereas the efficiency of high order self-associated species was reduced to less than 50%. Analysis of specific self-associated subclasses revealed that different factors influence the rate of HDL formation in vitro and ABCA1-mediated lipid release efficiency. The α-helix-forming ability of apoA-I is the main determinant of in vitro lipid solubilization rates, whereas loss of cellular lipid release efficiency is mainly caused by reduced structural flexibility by formation of stable quaternary interactions. Thus, stabilization of self-associated species impairs apoA-I biological activity through an ABCA1-mediated mechanism. These results afford mechanistic insights into the ABCA1 reaction and suggest self-association as a functional feature of apoA-I. Physiologic mechanisms may alter the native self-association state and contribute to apoA-I dysfunction.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Sumiko Abe-Dohmae
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Shinji Yokoyama
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Giorgio Cavigiolio
- Children's Hospital Oakland Research Institute, Oakland, California 94609.
| |
Collapse
|
39
|
Haase CL, Frikke-Schmidt R, Nordestgaard BG, Kateifides AK, Kardassis D, Nielsen LB, Andersen CB, Køber L, Johnsen AH, Grande P, Zannis VI, Tybjaerg-Hansen A. Mutation in APOA1 predicts increased risk of ischaemic heart disease and total mortality without low HDL cholesterol levels. J Intern Med 2011; 270:136-46. [PMID: 21443680 DOI: 10.1111/j.1365-2796.2011.02381.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To determine whether mutations in APOA1 affect levels of high-density lipoprotein (HDL) cholesterol and to predict risk of ischaemic heart disease (IHD) and total mortality in the general population. BACKGROUND Epidemiologically, risk of IHD is inversely related to HDL cholesterol levels. Mutations in apolipoprotein (apo) A-I, the major protein constituent of HDL, might be associated with low HDL cholesterol and predispose to IHD and early death. DESIGN We resequenced APOA1 in 190 individuals and examined the effect of mutations on HDL cholesterol, risk of IHD, myocardial infarction (MI) and mortality in 10 440 individuals in the prospective Copenhagen City Heart Study followed for 31 years. Results were validated in an independent case-control study (n = 16 035). Additionally, we determined plasma ratios of mutant to wildtype (WT) apoA-I in human heterozygotes and functional effects of mutations in adenovirus-transfected mice. RESULTS We identified a new mutation, A164S (1 : 500 in the general population), which predicted hazard ratios for IHD, MI and total mortality of 3.2 [95% confidence interval (CI): 1.6-6.5], 5.5 (95% CI: 2.6-11.7) and 2.5 (95% CI: 1.3-4.8), respectively, in heterozygotes compared with noncarriers. Mean reduction in survival time in heterozygotes was 10 years (P < 0.0001). Results for IHD and MI were confirmed in the case-control study. Furthermore, the ratio of mutant S164 to WT A164 apoA-I in plasma of heterozygotes was reduced. In addition, A164S heterozygotes had normal plasma lipid and lipoprotein levels, including HDL cholesterol and apoA-I, and this finding was confirmed in adenovirus-transfected mice. CONCLUSIONS A164S is the first mutation in APOA1 to be described that predicts an increased risk of IHD, MI and total mortality without low HDL cholesterol levels.
Collapse
Affiliation(s)
- C L Haase
- Department of Clinical Biochemistry, Rigshospitalet, Denmark
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ramella NA, Rimoldi OJ, Prieto ED, Schinella GR, Sanchez SA, Jaureguiberry MS, Vela ME, Ferreira ST, Tricerri MA. Human apolipoprotein A-I-derived amyloid: its association with atherosclerosis. PLoS One 2011; 6:e22532. [PMID: 21811627 PMCID: PMC3139661 DOI: 10.1371/journal.pone.0022532] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 06/23/2011] [Indexed: 01/08/2023] Open
Abstract
Amyloidoses constitute a group of diseases in which soluble proteins aggregate and deposit extracellularly in tissues. Nonhereditary apolipoprotein A-I (apoA-I) amyloid is characterized by deposits of nonvariant protein in atherosclerotic arteries. Despite being common, little is known about the pathogenesis and significance of apoA-I deposition. In this work we investigated by fluorescence and biochemical approaches the impact of a cellular microenvironment associated with chronic inflammation on the folding and pro-amyloidogenic processing of apoA-I. Results showed that mildly acidic pH promotes misfolding, aggregation, and increased binding of apoA-I to extracellular matrix elements, thus favoring protein deposition as amyloid like-complexes. In addition, activated neutrophils and oxidative/proteolytic cleavage of the protein give rise to pro amyloidogenic products. We conclude that, even though apoA-I is not inherently amyloidogenic, it may produce non hereditary amyloidosis as a consequence of the pro-inflammatory microenvironment associated to atherogenesis.
Collapse
Affiliation(s)
- Nahuel A. Ramella
- Instituto de Investigaciones Bioquímicas La Plata (INIBIOLP), CCT-CONICET, La Plata, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Omar J. Rimoldi
- Instituto de Investigaciones Bioquímicas La Plata (INIBIOLP), CCT-CONICET, La Plata, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Eduardo D. Prieto
- Instituto de Investigaciones Bioquímicas La Plata (INIBIOLP), CCT-CONICET, La Plata, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Guillermo R. Schinella
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Susana A. Sanchez
- Laboratory for Fluorescence Dynamics, University of California Irvine, Irvine, California, United States of America
- Microscopy Unit, Fundación CNIC-Carlos III, Centro Nacional de Investigaciones Cardiovasculares, Madrid, España
| | - María S. Jaureguiberry
- Instituto de Investigaciones Bioquímicas La Plata (INIBIOLP), CCT-CONICET, La Plata, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - María E. Vela
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Universidad Nacional de La Plata-CCT-CONICET, La Plata, Argentina
| | - Sergio T. Ferreira
- Program in Biochemistry and Cellular Biophysics, Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - M. Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas La Plata (INIBIOLP), CCT-CONICET, La Plata, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
41
|
Murphy C, Kestler D, Weiss D, Solomon A. Non-hereditary apolipoprotein AI-associated pulmonary amyloid. Amyloid 2011; 18 Suppl 1:219-20. [PMID: 21838494 DOI: 10.3109/13506129.2011.574354082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- C Murphy
- Human Immunology and Cancer Program, Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | | | | | | |
Collapse
|
42
|
|
43
|
Teoh CL, Bekard IB, Asimakis P, Griffin MDW, Ryan TM, Dunstan DE, Howlett GJ. Shear flow induced changes in apolipoprotein C-II conformation and amyloid fibril formation. Biochemistry 2011; 50:4046-57. [PMID: 21476595 DOI: 10.1021/bi2002482] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The misfolding and self-assembly of proteins into amyloid fibrils that occur in several debilitating diseases are affected by a variety of environmental factors, including mechanical factors associated with shear flow. We examined the effects of shear flow on amyloid fibril formation by human apolipoprotein C-II (apoC-II). Shear fields (150, 300, and 500 s(-1)) accelerated the rate of apoC-II fibril formation (1 mg/mL) approximately 5-10-fold. Fibrils produced at shear rates of 150 and 300 s(-1) were similar to the twisted ribbon fibrils formed in the absence of shear, while at 500 s(-1), tangled ropelike structures were observed. The mechanism of the shear-induced acceleration of amyloid fibril formation was investigated at low apoC-II concentrations (50 μg/mL) where fibril formation does not occur. Circular dichroism and tryptophan fluorescence indicated that shear induced an irreversible change in apoC-II secondary structure. Fluorescence resonance energy transfer experiments using the single tryptophan residue in apoC-II as the donor and covalently attached acceptors showed that shear flow increased the distance between the donor and acceptor molecules. Shear-induced higher-order oligomeric species were identified by sedimentation velocity experiments using fluorescence detection, while fibril seeding experiments showed that species formed during shear flow are on the fibril formation pathway. These studies suggest that physiological shear flow conditions and conditions experienced during protein manufacturing can exert significant effects on protein conformation, leading to protein misfolding, aggregation, and amyloid fibril formation.
Collapse
Affiliation(s)
- Chai Lean Teoh
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
44
|
Teoh CL, Griffin MDW, Howlett GJ. Apolipoproteins and amyloid fibril formation in atherosclerosis. Protein Cell 2011; 2:116-27. [PMID: 21400045 DOI: 10.1007/s13238-011-1013-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 01/29/2011] [Indexed: 10/18/2022] Open
Abstract
Amyloid fibrils arise from the aggregation of misfolded proteins into highly-ordered structures. The accumulation of these fibrils along with some non-fibrillar constituents within amyloid plaques is associated with the pathogenesis of several human degenerative diseases. A number of plasma apolipoproteins, including apolipoprotein (apo) A-I, apoA-II, apoC-II and apoE are implicated in amyloid formation or influence amyloid formation by other proteins. We review present knowledge of amyloid formation by apolipoproteins in disease, with particular focus on atherosclerosis. Further insights into the molecular mechanisms underlying their amyloidogenic propensity are obtained from in vitro studies which describe factors affecting apolipoprotein amyloid fibril formation and interactions. Additionally, we outline the evidence that amyloid fibril formation by apolipoproteins might play a role in the development and progression of atherosclerosis, and highlight possible molecular mechanisms that could contribute to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Chai Lean Teoh
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | |
Collapse
|
45
|
Dzwolak W. Vortex-induced chiral bifurcation in aggregating insulin. Chirality 2010; 22 Suppl 1:E154-60. [DOI: 10.1002/chir.20896] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 05/03/2010] [Indexed: 11/06/2022]
|
46
|
Smoak KA, Aloor JJ, Madenspacher J, Merrick BA, Collins JB, Zhu X, Cavigiolio G, Oda MN, Parks JS, Fessler MB. Myeloid differentiation primary response protein 88 couples reverse cholesterol transport to inflammation. Cell Metab 2010; 11:493-502. [PMID: 20519121 PMCID: PMC3091482 DOI: 10.1016/j.cmet.2010.04.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 02/03/2010] [Accepted: 04/05/2010] [Indexed: 02/06/2023]
Abstract
Crosstalk exists in mammalian cells between cholesterol trafficking and innate immune signaling. Apolipoprotein A-I (apoA-I), a serum apolipoprotein that induces antiatherogenic efflux of macrophage cholesterol, is widely described as anti-inflammatory because it neutralizes bacterial lipopolysaccharide. Conversely, lipopolysaccharide-induced inflammation is proatherogenic. However, whether innate immunity plays an endogenous, physiological role in host cholesterol homeostasis in the absence of infection is undetermined. We report that apoA-I signals in the macrophage through Toll-like receptor (TLR)2, TLR4, and CD14, utilizing myeloid differentiation primary response protein 88 (MyD88)-dependent and -independent pathways, to activate nuclear factor-kappaB and induce cytokines. MyD88 plays a critical role in reverse cholesterol transport in vitro and in vivo, in part through promoting ATP-binding cassette A1 transporter upregulation. Taken together, this work identifies apoA-I as an endogenous stimulus of innate immunity that couples cholesterol trafficking to inflammation through MyD88 and identifies innate immunity as a physiologic signal in cholesterol homeostasis.
Collapse
Affiliation(s)
- Kathleen A. Smoak
- Laboratory of Respiratory Biology, NIEHS, Research Triangle Park, NC 27709 U.S.A
| | - Jim J. Aloor
- Laboratory of Respiratory Biology, NIEHS, Research Triangle Park, NC 27709 U.S.A
| | | | - B. Alex Merrick
- Laboratory of Respiratory Biology, NIEHS, Research Triangle Park, NC 27709 U.S.A
| | | | - Xuewei Zhu
- Department of Pathology/Section on Lipid Sciences, Wake Forest University Health Sciences, Winston-Salem, NC 27157 U.S.A
| | | | - Michael N. Oda
- Children's Hospital Oakland Research Institute, Oakland, CA 94609 U.S.A
| | - John S. Parks
- Department of Pathology/Section on Lipid Sciences, Wake Forest University Health Sciences, Winston-Salem, NC 27157 U.S.A
| | - Michael B. Fessler
- Laboratory of Respiratory Biology, NIEHS, Research Triangle Park, NC 27709 U.S.A
| |
Collapse
|
47
|
Neyen C, Plüddemann A, Roversi P, Thomas B, Cai L, van der Westhuyzen DR, Sim RB, Gordon S. Macrophage scavenger receptor A mediates adhesion to apolipoproteins A-I and E. Biochemistry 2010; 48:11858-71. [PMID: 19911804 PMCID: PMC2793687 DOI: 10.1021/bi9013769] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Macrophage scavenger receptor A (SR-A) is a multifunctional, multiligand pattern recognition receptor with roles in innate immunity, apoptotic cell clearance, and age-related degenerative pathologies, such as atherosclerosis and Alzheimer's disease. Known endogenous SR-A ligands are polyanionic and include modified lipoproteins, advanced glycation end products, and extracellular matrix proteins. No native plasma ligands have been identified, but it is known that SR-A recognition of unidentified serum components mediates integrin-independent macrophage adhesion, which may drive chronic local inflammation. In this study, we used a high-throughput fractionation and screening method to identify novel endogenous SR-A ligands that may mediate macrophage adhesion. SR-A was found to recognize the exchangeable apolipoproteins A-I and E (apo A-I and apo E, respectively) in both lipid-free and lipid-associated form, suggesting the shared amphipathic alpha-helix as a potential recognition motif. Adhesion of RAW 264.7 macrophages to surfaces coated with apo A-I and apo E4 proved to be integrin-independent and could be blocked by anti-SR-A antibodies. The presence of apo A-I and apo E in pathological deposits, such as atherosclerotic lesions and neurotoxic Alzheimer's plaques, suggests a possible contribution of SR-A-dependent adhesion of macrophages to an inflammatory microenvironment.
Collapse
Affiliation(s)
- Claudine Neyen
- Sir William Dunn School of Pathology, Department of Biochemistry, University of Oxford,South Parks Road, Oxford OX13RE, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Methionine oxidation induces amyloid fibril formation by full-length apolipoprotein A-I. Proc Natl Acad Sci U S A 2010; 107:1977-82. [PMID: 20133843 DOI: 10.1073/pnas.0910136107] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Apolipoprotein A-I (apoA-I) is the major protein component of HDL, where it plays an important role in cholesterol transport. The deposition of apoA-I derived amyloid is associated with various hereditary systemic amyloidoses and atherosclerosis; however, very little is known about the mechanism of apoA-I amyloid formation. Methionine residues in apoA-I are oxidized via several mechanisms in vivo to form methionine sulfoxide (MetO), and significant levels of methionine oxidized apoA-I (MetO-apoA-I) are present in normal human serum. We investigated the effect of methionine oxidation on the structure, stability, and aggregation of full-length, lipid-free apoA-I. Circular dichrosim spectroscopy showed that oxidation of all three methionine residues in apoA-I caused partial unfolding of the protein and decreased its thermal stability, reducing the melting temperature (T(m)) from 58.7 degrees C for native apoA-I to 48.2 degrees C for MetO-apoA-I. Analytical ultracentrifugation revealed that methionine oxidation inhibited the native self association of apoA-I to form dimers and tetramers. Incubation of MetO-apoA-I for extended periods resulted in aggregation of the protein, and these aggregates bound Thioflavin T and Congo Red. Inspection of the aggregates by electron microscopy revealed fibrillar structures with a ribbon-like morphology, widths of approximately 11 nm, and lengths of up to several microns. X-ray fibre diffraction studies of the fibrils revealed a diffraction pattern with orthogonal peaks at spacings of 4.64 A and 9.92 A, indicating a cross-beta amyloid structure. This systematic study of fibril formation by full-length apoA-I represents the first demonstration that methionine oxidation can induce amyloid fibril formation.
Collapse
|
49
|
Loksztejn A, Dzwolak W. Vortex-Induced Formation of Insulin Amyloid Superstructures Probed by Time-Lapse Atomic Force Microscopy and Circular Dichroism Spectroscopy. J Mol Biol 2010; 395:643-55. [DOI: 10.1016/j.jmb.2009.10.065] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 10/25/2009] [Accepted: 10/27/2009] [Indexed: 11/16/2022]
|
50
|
Morita SY, Deharu Y, Takata E, Nakano M, Handa T. Cytotoxicity of lipid-free apolipoprotein B. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2008; 1778:2594-603. [DOI: 10.1016/j.bbamem.2008.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 08/19/2008] [Accepted: 08/19/2008] [Indexed: 11/17/2022]
|