1
|
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
|
2
|
Wang J, Li J, Zhong L. Current status and prospect of anti-amyloid fibril therapy in AL amyloidosis. Blood Rev 2024; 66:101207. [PMID: 38692939 DOI: 10.1016/j.blre.2024.101207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 05/03/2024]
Abstract
Amyloid light-chain (AL) amyloidosis is a rare hematological disease that produces abnormal monoclonal immunoglobulin light chains to form amyloid fibrils that are deposited in tissues, resulting in organ damage and dysfunction. Advanced AL amyloidosis has a very poor prognosis with a high risk of early mortality. The combination of anti-plasma cell therapy and amyloid fibrils clearance is the optimal treatment strategy, which takes into account both symptoms and root causes. However, research on anti-amyloid fibrils lags far behind research on anti-plasma cells, and there is currently no approved treatment that could clear amyloid fibrils. Nevertheless, anti-amyloid fibril therapies are being actively investigated recently and have shown potential in clinical trials. In this review, we aim to outline the preclinical work and clinical efficacy of fibril-directed therapies for AL amyloidosis.
Collapse
Affiliation(s)
- Jinghua Wang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jian Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Liye Zhong
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Wu D, Chen W. Molecular mechanisms and emerging therapies in wild-type transthyretin amyloid cardiomyopathy. Heart Fail Rev 2024; 29:511-521. [PMID: 38233673 PMCID: PMC10942909 DOI: 10.1007/s10741-023-10380-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/19/2024]
Abstract
Wild-type transthyretin amyloid cardiomyopathy (ATTRwt-CM) is an underrecognized cause of heart failure due to misfolded wild-type transthyretin (TTRwt) myocardial deposition. The development of wild-type TTR amyloid fibrils is a complex pathological process linked to the deterioration of homeostatic mechanisms owing to aging, plausibly implicating multiple molecular mechanisms. The components of amyloid transthyretin often include serum amyloid P, proteoglycans, and clusterin, which may play essential roles in the localization and elimination of amyloid fibrils. Oxidative stress, impaired mitochondrial function, and perturbation of intracellular calcium dynamics induced by TTR contribute to cardiac impairment. Recently, tafamidis has been the only drug approved by the U.S. Food and Drug Administration (FDA) for the treatment of ATTRwt-CM. In addition, small interfering RNAs and antisense oligonucleotides for ATTR-CM are promising therapeutic approaches and are currently in phase III clinical trials. Newly emerging therapies, such as antibodies targeting amyloid, inhibitors of seed formation, and CRISPR‒Cas9 technology, are currently in the early stages of research. The development of novel therapies is based on progress in comprehending the molecular events behind amyloid cardiomyopathy. There is still a need to further advance innovative treatments, providing patients with access to alternative and effective therapies, especially for patients diagnosed at a late stage.
Collapse
Affiliation(s)
- Danni Wu
- Dept. of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Wei Chen
- Dept. of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
4
|
Yang Z, Cao Y, Kong L, Xi J, Liu S, Zhang J, Cheng W. Small molecules as modulators of the proteostasis machinery: Implication in cardiovascular diseases. Eur J Med Chem 2024; 264:116030. [PMID: 38071793 DOI: 10.1016/j.ejmech.2023.116030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/25/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023]
Abstract
With the escalating prevalence of cardiovascular diseases, the substantial socioeconomic burden on healthcare systems is intensifying. Accumulating empirical evidence underscores the pivotal role of the proteostasis network in regulating cardiac homeostasis and function. Disruptions in proteostasis may contribute to the loss of protein function or the acquisition of toxic functions, which are intricately linked to the development of cardiovascular ailments such as atrial fibrillation, heart failure, atherosclerosis, and cardiac aging. It is widely acknowledged that the proteostasis network encompasses molecular chaperones, autophagy, and the ubiquitin proteasome system (UPS). Consequently, the proteostasis network emerges as an appealing target for therapeutic interventions in cardiovascular diseases. Numerous small molecules, acting as modulators of the proteostasis machinery, have exhibited therapeutic efficacy in managing cardiovascular diseases. This review centers on elucidating the role of the proteostasis network in various cardiovascular diseases and explores the potential of small molecules as therapeutic agents.
Collapse
Affiliation(s)
- Zhiheng Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yu Cao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Limin Kong
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Jianjun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Shourong Liu
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China.
| | - Jiankang Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Weiyan Cheng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Kovács P, Pushparaj PN, Takács R, Mobasheri A, Matta C. The clusterin connectome: Emerging players in chondrocyte biology and putative exploratory biomarkers of osteoarthritis. Front Immunol 2023; 14:1103097. [PMID: 37033956 PMCID: PMC10081159 DOI: 10.3389/fimmu.2023.1103097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
IntroductionClusterin is amoonlighting protein that hasmany functions. It is amultifunctional Q6 holdase chaperone glycoprotein that is present intracellularly and extracellularly in almost all bodily fluids. Clusterin is involved in lipid transport, cell differentiation, regulation of apoptosis, and clearance of cellular debris, and plays a protective role in ensuring cellular survival. However, the possible involvement of clusterin in arthritic disease remains unclear. Given the significant potential of clusterin as a biomarker of osteoarthritis (OA), a more detailed analysis of its complex network in an inflammatory environment, specifically in the context of OA, is required. Based on the molecular network of clusterin, this study aimed to identify interacting partners that could be developed into biomarker panels for OA.MethodsThe STRING database and Cytoscape were used to map and visualize the clusterin connectome. The Qiagen Ingenuity Pathway Analysis (IPA) software was used to analyze and study clusterinassociated signaling networks in OA. We also analyzed transcription factors known to modulate clusterin expression, which may be altered in OA.ResultsThe top hits in the clusterin network were intracellular chaperones, aggregate-forming proteins, apoptosis regulators and complement proteins. Using a text-mining approach in Cytoscape, we identified additional interacting partners, including serum proteins, apolipoproteins, and heat shock proteins.DiscussionBased on known interactions with proteins, we predicted potential novel components of the clusterin connectome in OA, including selenoprotein R, semaphorins, and meprins, which may be important for designing new prognostic or diagnostic biomarker panels.
Collapse
Affiliation(s)
- Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research (CEGMR), Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ali Mobasheri
- FibroHealth Interdisciplinary Research Programme, Fibrobesity Cluster, Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
- *Correspondence: Csaba Matta, ; Ali Mobasheri,
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- *Correspondence: Csaba Matta, ; Ali Mobasheri,
| |
Collapse
|
6
|
Misumi Y, Tabata Y, Tasaki M, Obayashi K, Yamakawa S, Nomura T, Ueda M. Binding of serum-derived amyloid-associated proteins to amyloid fibrils. Amyloid 2023; 30:67-73. [PMID: 36094798 DOI: 10.1080/13506129.2022.2120800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Amyloid signature proteins such as serum amyloid P component, apolipoprotein E (ApoE), and ApoA-IV generally co-localise with amyloid, regardless of the types of amyloid precursor protein or the organs. Most of these proteins derive from serum and have reportedly been involved in amyloid fibril formation and stabilisation, as well as in excretion and degradation of amyloid precursor proteins. However, the processes and mechanisms by which these specific proteins deposit together with amyloid fibrils have not been clarified. METHODS We analysed the binding of serum proteins to amyloid fibrils derived from amyloid β and insulin in vitro by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS Specific serum proteins including ApoA-I, ApoE, ApoA-IV, ApoC-III and vitronectin adhered to amyloid fibrils at high concentrations in vitro. In addition, the profile of these proteins commonly occurred in both amyloid β and insulin amyloid fibrils and was mostly consistent with the composition of amyloid signature proteins. We also showed that high concentrations of serum proteins can adhere to amyloid fibrils in a short time. CONCLUSIONS Our in vitro results suggest that amyloid signature proteins coexist with amyloid primarily dependent on the binding of each serum protein, in the extracellular fluid, to amyloid fibrils.
Collapse
Affiliation(s)
- Yohei Misumi
- Department of Neurology, Kumamoto University, Kumamoto, Japan
| | - Yuri Tabata
- Department of Neurology, Kumamoto University, Kumamoto, Japan
| | - Masayoshi Tasaki
- Department of Neurology, Kumamoto University, Kumamoto, Japan.,Department of Biomedical Laboratory Sciences, Kumamoto University, Kumamoto, Japan
| | - Konen Obayashi
- Department of Morphological and Physiological Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shiori Yamakawa
- Department of Neurology, Kumamoto University, Kumamoto, Japan
| | - Toshiya Nomura
- Department of Neurology, Kumamoto University, Kumamoto, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
7
|
Absmeier RM, Rottenaicher GJ, Svilenov HL, Kazman P, Buchner J. Antibodies gone bad - the molecular mechanism of light chain amyloidosis. FEBS J 2023; 290:1398-1419. [PMID: 35122394 DOI: 10.1111/febs.16390] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/19/2022] [Accepted: 02/03/2022] [Indexed: 12/19/2022]
Abstract
Light chain amyloidosis (AL) is a systemic disease in which abnormally proliferating plasma cells secrete large amounts of mutated antibody light chains (LCs) that eventually form fibrils. The fibrils are deposited in various organs, most often in the heart and kidney, and impair their function. The prognosis for patients diagnosed with AL is generally poor. The disease is set apart from other amyloidoses by the huge number of patient-specific mutations in the disease-causing and fibril-forming protein. The molecular mechanisms that drive the aggregation of mutated LCs into fibrils have been enigmatic, which hindered the development of efficient diagnostics and therapies. In this review, we summarize our current knowledge on AL amyloidosis and discuss open issues.
Collapse
Affiliation(s)
- Ramona M Absmeier
- Center for Functional Protein Assemblies and Department of Chemistry, Technische Universität München, Garching, Germany
| | - Georg J Rottenaicher
- Center for Functional Protein Assemblies and Department of Chemistry, Technische Universität München, Garching, Germany
| | - Hristo L Svilenov
- Center for Functional Protein Assemblies and Department of Chemistry, Technische Universität München, Garching, Germany
| | - Pamina Kazman
- Center for Functional Protein Assemblies and Department of Chemistry, Technische Universität München, Garching, Germany
| | - Johannes Buchner
- Center for Functional Protein Assemblies and Department of Chemistry, Technische Universität München, Garching, Germany
| |
Collapse
|
8
|
Martinez-Rivas G, Bender S, Sirac C. Understanding AL amyloidosis with a little help from in vivo models. Front Immunol 2022; 13:1008449. [PMID: 36458006 PMCID: PMC9707859 DOI: 10.3389/fimmu.2022.1008449] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/27/2022] [Indexed: 08/01/2023] Open
Abstract
Monoclonal immunoglobulin (Ig) light chain amyloidosis (AL) is a rare but severe disease that may occur when a B or plasma cell clone secretes an excess of free Ig light chains (LCs). Some of these LCs tend to aggregate into organized fibrils with a β-sheet structure, the so-called amyloid fibrils, and deposit into the extracellular compartment of organs, such as the heart or kidneys, causing their dysfunction. Recent findings have confirmed that the core of the amyloid fibrils is constituted by the variable (V) domain of the LCs, but the mechanisms underlying the unfolding and aggregation of this fragment and its deposition are still unclear. Moreover, in addition to the mechanical constraints exerted by the massive accumulation of amyloid fibrils in organs, the direct toxicity of these variable domain LCs, full-length light chains, or primary amyloid precursors (oligomers) seems to play a role in the pathogenesis of the disease. Many in vitro studies have focused on these topics, but the variability of this disease, in which each LC presents unique properties, and the extent and complexity of affected organs make its study in vivo very difficult. Accordingly, several groups have focused on the development of animal models for years, with some encouraging but mostly disappointing results. In this review, we discuss the experimental models that have been used to better understand the unknowns of this pathology with an emphasis on in vivo approaches. We also focus on why reliable AL amyloidosis animal models remain so difficult to obtain and what this tells us about the pathophysiology of the disease.
Collapse
|
9
|
Ikura H, Endo J, Kitakata H, Moriyama H, Sano M, Fukuda K. Molecular Mechanism of Pathogenesis and Treatment Strategies for AL Amyloidosis. Int J Mol Sci 2022; 23:6336. [PMID: 35683015 PMCID: PMC9181426 DOI: 10.3390/ijms23116336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 11/17/2022] Open
Abstract
In amyloid light-chain (AL) amyloidosis, small B-cell clones (mostly plasma cell clones) present in the bone marrow proliferate and secrete unstable monoclonal free light chains (FLCs), which form amyloid fibrils that deposit in the interstitial tissue, resulting in organ injury and dysfunction. AL amyloidosis progresses much faster than other types of amyloidosis, with a slight delay in diagnosis leading to a marked exacerbation of cardiomyopathy. In some cases, the resulting heart failure is so severe that chemotherapy cannot be administered, and death sometimes occurs within a few months. To date, many clinical studies have focused on therapeutics, especially chemotherapy, to treat this disease. Because it is necessary to promptly lower FLC, the causative protein of amyloid, to achieve a hematological response, various anticancer agents targeting neoplastic plasma cells are used for the treatment of this disease. In addition, many basic studies using human specimens to elucidate the pathophysiology of AL have been conducted. Gene mutations associated with AL, the characteristics of amyloidogenic LC, and the structural specificity of amyloid fibrils have been clarified. Regarding the mechanism of cellular and tissue damage, the mass effect due to amyloid deposition, as well as the toxicity of pre-fibrillar LC, is gradually being elucidated. This review outlines the pathogenesis and treatment strategies for AL amyloidosis with respect to its molecular mechanisms.
Collapse
Affiliation(s)
| | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-City, Tokyo 160-8582, Japan; (H.I.); (H.K.); (H.M.); (M.S.); (K.F.)
| | | | | | | | | |
Collapse
|
10
|
Du R, Liu G, Huang H. 2-Hydroxyisobutyrylome in Mouse Liver Expands the Roles of Lysine 2-hydroxyisobutyrylation Pathway. Bioorg Med Chem 2022; 57:116634. [DOI: 10.1016/j.bmc.2022.116634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
|
11
|
Palstrøm NB, Rojek AM, Møller HEH, Hansen CT, Matthiesen R, Rasmussen LM, Abildgaard N, Beck HC. Classification of Amyloidosis by Model-Assisted Mass Spectrometry-Based Proteomics. Int J Mol Sci 2021; 23:ijms23010319. [PMID: 35008745 PMCID: PMC8745254 DOI: 10.3390/ijms23010319] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 11/16/2022] Open
Abstract
Amyloidosis is a rare disease caused by the misfolding and extracellular aggregation of proteins as insoluble fibrillary deposits localized either in specific organs or systemically throughout the body. The organ targeted and the disease progression and outcome is highly dependent on the specific fibril-forming protein, and its accurate identification is essential to the choice of treatment. Mass spectrometry-based proteomics has become the method of choice for the identification of the amyloidogenic protein. Regrettably, this identification relies on manual and subjective interpretation of mass spectrometry data by an expert, which is undesirable and may bias diagnosis. To circumvent this, we developed a statistical model-assisted method for the unbiased identification of amyloid-containing biopsies and amyloidosis subtyping. Based on data from mass spectrometric analysis of amyloid-containing biopsies and corresponding controls. A Boruta method applied on a random forest classifier was applied to proteomics data obtained from the mass spectrometric analysis of 75 laser dissected Congo Red positive amyloid-containing biopsies and 78 Congo Red negative biopsies to identify novel “amyloid signature” proteins that included clusterin, fibulin-1, vitronectin complement component C9 and also three collagen proteins, as well as the well-known amyloid signature proteins apolipoprotein E, apolipoprotein A4, and serum amyloid P. A SVM learning algorithm were trained on the mass spectrometry data from the analysis of the 75 amyloid-containing biopsies and 78 amyloid-negative control biopsies. The trained algorithm performed superior in the discrimination of amyloid-containing biopsies from controls, with an accuracy of 1.0 when applied to a blinded mass spectrometry validation data set of 103 prospectively collected amyloid-containing biopsies. Moreover, our method successfully classified amyloidosis patients according to the subtype in 102 out of 103 blinded cases. Collectively, our model-assisted approach identified novel amyloid-associated proteins and demonstrated the use of mass spectrometry-based data in clinical diagnostics of disease by the unbiased and reliable model-assisted classification of amyloid deposits and of the specific amyloid subtype.
Collapse
Affiliation(s)
- Nicolai Bjødstrup Palstrøm
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Aleksandra M. Rojek
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
| | - Hanne E. H. Møller
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
| | - Charlotte Toftmann Hansen
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Department of Hematology, Odense University Hospital, 5000 Odense, Denmark
| | - Rune Matthiesen
- Computational and Experimental Biology Group, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal;
| | - Lars Melholt Rasmussen
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
| | - Niels Abildgaard
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Department of Hematology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
- Haematology Pathology Research Laboratory, Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
| | - Hans Christian Beck
- Odense Amyloidosis Center, Odense University Hospital, 5000 Odense, Denmark; (N.B.P.); (A.M.R.); (H.E.H.M.); (C.T.H.); (L.M.R.); (N.A.)
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
- Correspondence: ; Tel.: +45-29647470
| |
Collapse
|
12
|
Obici L, Mussinelli R. Current and Emerging Therapies for Hereditary Transthyretin Amyloidosis: Strides Towards a Brighter Future. Neurotherapeutics 2021; 18:2286-2302. [PMID: 34850359 PMCID: PMC8804119 DOI: 10.1007/s13311-021-01154-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 12/19/2022] Open
Abstract
The past few years have witnessed an unprecedented acceleration in the clinical development of novel therapeutic options for hereditary transthyretin amyloidosis. Recently approved agents and drugs currently under investigation not only represent a major breakthrough in this field but also provide validation of the therapeutic potential of innovative approaches, like RNA interference and CRISPR-Cas9-mediated gene editing, in rare inherited disorders. In this review, we describe the evolving therapeutic landscape for hereditary transthyretin amyloidosis and discuss how this highly disabling and fatal condition is turning into a treatable disease. We also provide an overview of the molecular mechanisms involved in transthyretin (TTR) amyloid formation and regression, to highlight how a deeper understanding of these processes has contributed to the identification of novel treatment targets. Finally, we focus on major areas of uncertainty and unmet needs that deserve further efforts to improve long-term patients' outcomes and allow for a brighter future.
Collapse
Affiliation(s)
- Laura Obici
- Amyloidosis Research and Treatment Centre, IRCCS Fondazione Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy.
| | - Roberta Mussinelli
- Amyloidosis Research and Treatment Centre, IRCCS Fondazione Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy
| |
Collapse
|
13
|
Rostagno A, Calero M, Holton JL, Revesz T, Lashley T, Ghiso J. Association of clusterin with the BRI2-derived amyloid molecules ABri and ADan. Neurobiol Dis 2021; 158:105452. [PMID: 34298087 PMCID: PMC8440498 DOI: 10.1016/j.nbd.2021.105452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 06/30/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022] Open
Abstract
Familial British and Danish dementias (FBD and FDD) share striking neuropathological similarities with Alzheimer's disease (AD), including intraneuronal neurofibrillary tangles as well as parenchymal and vascular amyloid deposits. Multiple amyloid associated proteins with still controversial role in amyloidogenesis colocalize with the structurally different amyloid peptides ABri in FBD, ADan in FDD, and Aβ in AD. Genetic variants and plasma levels of one of these associated proteins, clusterin, have been identified as risk factors for AD. Clusterin is known to bind soluble Aβ in biological fluids, facilitate its brain clearance, and prevent its aggregation. The current work identifies clusterin as the major ABri- and ADan-binding protein and provides insight into the biochemical mechanisms leading to the association of clusterin with ABri and ADan deposits. Mirroring findings in AD, the studies corroborate clusterin co-localization with cerebral parenchymal and vascular amyloid deposits in both disorders. Ligand affinity chromatography with downstream Western blot and amino acid sequence analyses unequivocally identified clusterin as the major ABri- and ADan-binding plasma protein. ELISA highlighted a specific saturable binding of clusterin to ABri and ADan with low nanomolar Kd values within the same range as those previously demonstrated for the clusterin-Aβ interaction. Consistent with its chaperone activity, thioflavin T binding assays clearly showed a modulatory effect of clusterin on ABri and ADan aggregation/fibrillization properties. Our findings, together with the known multifunctional activity of clusterin and its modulatory activity on the complex cellular pathways leading to oxidative stress, mitochondrial dysfunction, and the induction of cell death mechanisms - all known pathogenic features of these protein folding disorders - suggests the likelihood of a more complex role and a translational potential for the apolipoprotein in the amelioration/prevention of these pathogenic mechanisms.
Collapse
Affiliation(s)
- Agueda Rostagno
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Miguel Calero
- Instituto de Salud Carlos III, 28029 Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; Alzheimer's Center Reina Sofia Foundation - CIEN Foundation, 28031 Madrid, Spain
| | - Janice L Holton
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Tamas Revesz
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Tammaryn Lashley
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Jorge Ghiso
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
14
|
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: 23] [Impact Index Per Article: 5.8] [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
|
15
|
Gottwald J, Röcken C. The amyloid proteome: a systematic review and proposal of a protein classification system. Crit Rev Biochem Mol Biol 2021; 56:526-542. [PMID: 34311636 DOI: 10.1080/10409238.2021.1937926] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyloidosis is a disease caused by pathological fibril aggregation and deposition of proteins in different tissues and organs. Thirty-six fibril-forming proteins have been identified. So far, proteomic evaluation of amyloid focused on the detection and characterization of fibril proteins mainly for diagnostic purposes or to find novel fibril-forming proteins. However, amyloid deposits are a complex mixture of constituents that show organ-, tissue-, and amyloid-type specific patterns, that is the amyloid proteome. We carried out a comprehensive literature review on publications investigating amyloid via liquid chromatography coupled to tandem mass spectrometry, including but not limited to sample preparation by laser microdissection. Our review confirms the complexity and dynamics of the amyloid proteome, which can be divided into four functional categories: amyloid proteome-category 1 (APC1) includes exclusively fibrillary proteins found in the patient; APC2 includes potential fibril-forming proteins found in other types of amyloid; and APC3 and APC4 summarizes non-fibril proteins-some being amyloid signature proteins. Our categorization may help to systemically explore the nature and role of the amyloid proteome in the manifestation, progression, and clearance of disease. Further exploration of the amyloid proteome may form the basis for the development of novel diagnostic tools, thereby enabling the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Juliane Gottwald
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
16
|
Bezerra F, Saraiva MJ, Almeida MR. Modulation of the Mechanisms Driving Transthyretin Amyloidosis. Front Mol Neurosci 2020; 13:592644. [PMID: 33362465 PMCID: PMC7759661 DOI: 10.3389/fnmol.2020.592644] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022] Open
Abstract
Transthyretin (TTR) amyloidoses are systemic diseases associated with TTR aggregation and extracellular deposition in tissues as amyloid. The most frequent and severe forms of the disease are hereditary and associated with amino acid substitutions in the protein due to single point mutations in the TTR gene (ATTRv amyloidosis). However, the wild type TTR (TTR wt) has an intrinsic amyloidogenic potential that, in particular altered physiologic conditions and aging, leads to TTR aggregation in people over 80 years old being responsible for the non-hereditary ATTRwt amyloidosis. In normal physiologic conditions TTR wt occurs as a tetramer of identical subunits forming a central hydrophobic channel where small molecules can bind as is the case of the natural ligand thyroxine (T4). However, the TTR amyloidogenic variants present decreased stability, and in particular conditions, dissociate into partially misfolded monomers that aggregate and polymerize as amyloid fibrils. Therefore, therapeutic strategies for these amyloidoses may target different steps in the disease process such as decrease of variant TTR (TTRv) in plasma, stabilization of TTR, inhibition of TTR aggregation and polymerization or disruption of the preformed fibrils. While strategies aiming decrease of the mutated TTR involve mainly genetic approaches, either by liver transplant or the more recent technologies using specific oligonucleotides or silencing RNA, the other steps of the amyloidogenic cascade might be impaired by pharmacologic compounds, namely, TTR stabilizers, inhibitors of aggregation and amyloid disruptors. Modulation of different steps involved in the mechanism of ATTR amyloidosis and compounds proposed as pharmacologic agents to treat TTR amyloidosis will be reviewed and discussed.
Collapse
Affiliation(s)
- Filipa Bezerra
- Molecular Neurobiology Group, IBMC-Instituto de Biologia Molecular e Celular, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal.,Department of Molecular Biology, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria João Saraiva
- Molecular Neurobiology Group, IBMC-Instituto de Biologia Molecular e Celular, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal.,Department of Molecular Biology, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Rosário Almeida
- Molecular Neurobiology Group, IBMC-Instituto de Biologia Molecular e Celular, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal.,Department of Molecular Biology, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
17
|
Torres-Arancivia CM, Chang D, Hackett WE, Zaia J, Connors LH. Glycosylation of Serum Clusterin in Wild-Type Transthyretin-Associated (ATTRwt) Amyloidosis: A Study of Disease-Associated Compositional Features Using Mass Spectrometry Analyses. Biochemistry 2020; 59:4367-4378. [PMID: 33141553 PMCID: PMC8082438 DOI: 10.1021/acs.biochem.0c00590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Wild-type transthyretin-associated (ATTRwt) amyloidosis is an age-related disease that causes heart failure in older adults. This disease frequently features cardiac amyloid fibril deposits that originate from dissociation of the tetrameric protein, transthyretin (TTR). Unlike hereditary TTR (ATTRm) amyloidosis, where amino acid replacements destabilize the native protein, in ATTRwt amyloidosis, amyloid-forming TTR lacks protein sequence alterations. The initiating cause of fibril formation in ATTRwt amyloidosis is unclear, and thus, it seems plausible that other factors are involved in TTR misfolding and unregulated accumulation of wild-type TTR fibrils. We believe that clusterin (CLU, UniProtKB P10909), a plasma circulating glycoprotein, plays a role in the pathobiology of ATTRwt amyloidosis. Previously, we have suggested a role for CLU in ATTRwt amyloidosis based on our studies showing that (1) CLU codeposits with non-native TTR in amyloid fibrils from ATTRwt cardiac tissue, (2) CLU interacts only with non-native (monomeric and aggregated) forms of TTR, and (3) CLU serum levels in patients with ATTRwt are significantly lower compared to healthy controls. In the present study, we provide comprehensive detail of compositional findings from mass spectrometry analyses of amino acid and glycan content of CLU purified from ATTRwt and control sera. The characterization of oligosaccharide content in serum CLU derived from patients with ATTRwt amyloidosis is novel data. Moreover, results comparing CLU oligosaccharide variations between patient and healthy controls are original and provide further evidence for the role of CLU in ATTRwt pathobiology, possibly linked to disease-specific structural features that limit the chaperoning capacity of CLU.
Collapse
|
18
|
Abah SE, Burté F, Howell SA, Lagunju I, Shokunbi WA, Wahlgren M, Sodeinde O, Brown BJ, Holder AA, Fernandez-Reyes D. Depleted circulatory complement-lysis inhibitor (CLI) in childhood cerebral malaria returns to normal with convalescence. Malar J 2020; 19:167. [PMID: 32336276 PMCID: PMC7184698 DOI: 10.1186/s12936-020-03241-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/17/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Cerebral malaria (CM), is a life-threatening childhood malaria syndrome with high mortality. CM is associated with impaired consciousness and neurological damage. It is not fully understood, as yet, why some children develop CM. Presented here is an observation from longitudinal studies on CM in a paediatric cohort of children from a large, densely-populated and malaria holoendemic, sub-Saharan, West African metropolis. METHODS Plasma samples were collected from a cohort of children with CM, severe malarial anaemia (SMA), uncomplicated malaria (UM), non-malaria positive healthy community controls (CC), and coma and anemic patients without malaria, as disease controls (DC). Proteomic two-dimensional difference gel electrophoresis (2D-DIGE) and mass spectrometry were used in a discovery cohort to identify plasma proteins that might be discriminatory among these clinical groups. The circulatory levels of identified proteins of interest were quantified by ELISA in a prospective validation cohort. RESULTS The proteome analysis revealed differential abundance of circulatory complement-lysis inhibitor (CLI), also known as Clusterin (CLU). CLI circulatory level was low at hospital admission in all children presenting with CM and recovered to normal level during convalescence (p < 0.0001). At acute onset, circulatory level of CLI in the CM group significantly discriminates CM from the UM, SMA, DC and CC groups. CONCLUSIONS The CLI circulatory level is low in all patients in the CM group at admission, but recovers through convalescence. The level of CLI at acute onset may be a specific discriminatory marker of CM. This work suggests that CLI may play a role in the pathophysiology of CM and may be useful in the diagnosis and follow-up of children presenting with CM.
Collapse
Affiliation(s)
| | - Florence Burté
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Steven A Howell
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Ikeoluwa Lagunju
- Department of Paediatrics, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
- Childhood Malaria Research Group, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
| | - Wuraola A Shokunbi
- Childhood Malaria Research Group, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
- Department of Haematology, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
| | - Mats Wahlgren
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Olugbemiro Sodeinde
- Department of Paediatrics, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
- Childhood Malaria Research Group, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
- Department of Computer Science, Faculty of Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Biobele J Brown
- Department of Paediatrics, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
- Childhood Malaria Research Group, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria
| | | | - Delmiro Fernandez-Reyes
- Department of Paediatrics, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria.
- Childhood Malaria Research Group, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria.
- Department of Computer Science, Faculty of Engineering, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
19
|
Affiliation(s)
| | - Lawreen H Connors
- a Amyloidosis Center, Boston University School of Medicine , Boston , MA, USA
| |
Collapse
|
20
|
Abstract
The heart, like any organ in the body, is susceptible to amyloid deposition. Although more than 30 types of protein can cause amyloidosis, only two types commonly deposit in the ventricular myocardium: amyloid light chain and amyloid transthyretin. Amyloid cardiomyopathy is usually a major determinant of patient outcomes, and the diagnosis of heart involvement can be often relatively under-diagnosed, owing to nonspecific presenting symptoms and signs at a subclinical stage. The diagnosis of cardiac amyloidosis is usually performed by endomyocardial biopsy; however, the invasive nature and related high-risk complications restrict its wide use in clinical settings. Recently, with the advent of innovative techniques used for evaluating cardiac amyloidosis, noninvasive methods become increasingly important, especially in earlier diagnosis, distinguishing typing, risk prediction and response to treatment. Here, we will review recent developments in the noninvasive methods used in the assessment of cardiac amyloidosis, focused on the laboratory biomarkers and imaging modalities.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Dongcheng district, Beijing, 100730, China
| | - Quan Fang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Dongcheng district, Beijing, 100730, China.
| |
Collapse
|
21
|
Gregory JM, Whiten DR, Brown RA, Barros TP, Kumita JR, Yerbury JJ, Satapathy S, McDade K, Smith C, Luheshi LM, Dobson CM, Wilson MR. Clusterin protects neurons against intracellular proteotoxicity. Acta Neuropathol Commun 2017; 5:81. [PMID: 29115989 PMCID: PMC5678579 DOI: 10.1186/s40478-017-0481-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 12/11/2022] Open
Abstract
It is now widely accepted in the field that the normally secreted chaperone clusterin is redirected to the cytosol during endoplasmic reticulum (ER) stress, although the physiological function(s) of this physical relocation remain unknown. We have examined in this study whether or not increased expression of clusterin is able to protect neuronal cells against intracellular protein aggregation and cytotoxicity, characteristics that are strongly implicated in a range of neurodegenerative diseases. We used the amyotrophic lateral sclerosis-associated protein TDP-43 as a primary model to investigate the effects of clusterin on protein aggregation and neurotoxicity in complementary in vitro, neuronal cell and Drosophila systems. We have shown that clusterin directly interacts with TDP-43 in vitro and potently inhibits its aggregation, and observed that in ER stressed neuronal cells, clusterin co-localized with TDP-43 and specifically reduced the numbers of cytoplasmic inclusions. We further showed that the expression of TDP-43 in transgenic Drosophila neurons induced ER stress and that co-expression of clusterin resulted in a dramatic clearance of mislocalized TDP-43 from motor neuron axons, partially rescued locomotor activity and significantly extended lifespan. We also showed that in Drosophila photoreceptor cells, clusterin co-expression gave ER stress-dependent protection against proteotoxicity arising from both Huntingtin-Q128 and mutant (R406W) human tau. We therefore conclude that increased expression of clusterin can provide an important defense against intracellular proteotoxicity under conditions that mimic specific features of neurodegenerative disease.
Collapse
Affiliation(s)
- Jenna M Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, 49 Little France Crescent-Chancellor, Edinburgh, EH16 4SB, UK
| | - Daniel R Whiten
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Rebecca A Brown
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Teresa P Barros
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Janet R Kumita
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Sandeep Satapathy
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Karina McDade
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Leila M Luheshi
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
22
|
Bezerra Júnior RQ, Eloy ÂMX, Furtado JR, Pinheiro RR, Andrioli A, Moreno FB, Pinto Lobo MD, Monteiro-Moreira ACO, de Azevedo Moreira R, Farias Pinto TM, da Silva Teixeira MF. A panel of protein candidates for comprehensive study of Caprine Arthritis Encephalitis (CAE) infection. Trop Anim Health Prod 2017; 50:43-48. [DOI: 10.1007/s11250-017-1398-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 09/05/2017] [Indexed: 01/06/2023]
|
23
|
Clusterin expression in medullary thyroid carcinoma is inversely correlated with the presence of lymph node metastases. Hum Pathol 2017; 64:37-43. [DOI: 10.1016/j.humpath.2017.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/06/2017] [Accepted: 03/19/2017] [Indexed: 12/21/2022]
|
24
|
Lavatelli F, Merlini G. Advances in proteomic study of cardiac amyloidosis: progress and potential. Expert Rev Proteomics 2016; 13:1017-1027. [PMID: 27678147 DOI: 10.1080/14789450.2016.1242417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION More than ten distinct forms of amyloidoses that can involve the heart have been described, classified according to which protein originates the deposits. Cardiac amyloid infiltration translates into progressive and often life-threatening cardiomyopathy, but disease severity, prognosis and treatment drastically differ according to the amyloidosis type. The notion that protein misfolding and aggregation play a more general role in human cardiomyopathies has further raised attention towards the definition of the proteotoxicity mechanisms. Areas covered: Mass spectrometry-based proteomics plays an important role as a diagnostic tool and for understanding the molecular bases of amyloid cardiomyopathies. The landscape of applications of proteomics to the study of cardiac amyloidoses and amyloid-related cardiotoxicity is summarized, with a critical synthesis of the major achievements. Expert commentary: Current strengths and limitations of proteomics in the clinical setting and in translational research on amyloid cardiomyopathy are discussed, with the foreseen potential future directions in the field.
Collapse
Affiliation(s)
- Francesca Lavatelli
- a Amyloidosis Research and Treatment Center , Fondazione IRCCS Policlinico San Matteo, and University of Pavia , Pavia , Italy
| | - Giampaolo Merlini
- a Amyloidosis Research and Treatment Center , Fondazione IRCCS Policlinico San Matteo, and University of Pavia , Pavia , Italy
| |
Collapse
|
25
|
Panayiotou E, Papacharalambous R, Antoniou A, Christophides G, Papageorgiou L, Fella E, Malas S, Kyriakides T. Genetic background modifies amyloidosis in a mouse model of ATTR neuropathy. Biochem Biophys Rep 2016; 8:48-54. [PMID: 28955941 PMCID: PMC5613746 DOI: 10.1016/j.bbrep.2016.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/04/2016] [Accepted: 08/04/2016] [Indexed: 11/29/2022] Open
Abstract
Penetrance and age of onset of ATTRV30M amyloidotic neuropathy varies significantly among different populations. This variability has been attributed to both genetic and environmental modifiers. We studied the effect of genetic background on phenotype in two lines of transgenic mice bearing the same ATTRV30M transgene. Amyloid deposition, transthyretin (TTR), megalin, clusterin and disease markers of endoplasmic reticulum stress, the ubiquitin-proteasome system, apoptosis, and complement activation were assessed with WB and immunohistochemistry in donor and recipient tissue. Our results indicate that genetic background modulates amyloid deposition by influencing TTR handling in recipient tissue and may partly account for the marked variability in penetrance observed in various world populations. Genetic background modulates ATTR amyloid deposition. Genetic background affects pathogenic cascades involved in amyloidogenesis. Megalin and clustering possibly involved in the handing of TTR monomers.
Collapse
Affiliation(s)
- E Panayiotou
- Cyprus Institute of Neurology and Genetics, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - R Papacharalambous
- Cyprus Institute of Neurology and Genetics, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - A Antoniou
- Cyprus School of Molecular Medicine, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - G Christophides
- Cyprus School of Molecular Medicine, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - L Papageorgiou
- Cyprus School of Molecular Medicine, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - E Fella
- Cyprus School of Molecular Medicine, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - S Malas
- Cyprus Institute of Neurology and Genetics, P.O.Box 23462, 1683 Nicosia, Cyprus
| | - T Kyriakides
- Cyprus Institute of Neurology and Genetics, P.O.Box 23462, 1683 Nicosia, Cyprus.,Cyprus School of Molecular Medicine, P.O.Box 23462, 1683 Nicosia, Cyprus
| |
Collapse
|
26
|
The role of fibrinogen glycation in ATTR: evidence for chaperone activity loss in disease. Biochem J 2016; 473:2225-37. [DOI: 10.1042/bcj20160290] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 05/12/2016] [Indexed: 12/14/2022]
Abstract
Transthyretin amyloidosis (ATTR) belongs to a class of disorders caused by protein misfolding and aggregation. ATTR is a disabling disorder of autosomal dominant trait, where transthyretin (TTR) forms amyloid deposits in different organs, causing dysfunction of the peripheral nervous system. We previously discovered that amyloid fibrils from ATTR patients are glycated by methylglyoxal. Even though no consensus has been reached about the actual role of methylglyoxal-derived advanced glycation end-products in amyloid diseases, evidence collected so far points to a role for protein glycation in conformational abnormalities, being ubiquitously found in amyloid deposits in Alzheimer's disease, dialysis-related amyloidosis and Parkinson's diseases. Human fibrinogen, an extracellular chaperone, was reported to specifically interact with a wide spectrum of stressed proteins and suppress their aggregation, being an interacting protein with TTR. Fibrinogen is differentially glycated in ATTR, leading to its chaperone activity loss. Here we show the existence of a proteostasis imbalance in ATTR linked to fibrinogen glycation by methylglyoxal.
Collapse
|
27
|
Genereux JC, Wiseman RL. Regulating extracellular proteostasis capacity through the unfolded protein response. Prion 2016; 9:10-21. [PMID: 25946012 DOI: 10.1080/19336896.2015.1011887] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The extracellular aggregation of proteins into proteotoxic oligomers and amyloid fibrils is implicated in the onset and pathology of numerous diseases referred to as amyloid diseases. All of the proteins that aggregate extracellularly in association with amyloid disease pathogenesis originate in the endoplasmic reticulum (ER) and are secreted through the secretory pathway. Disruptions in ER protein homeostasis or proteostasis (i.e., ER stress) can facilitate the aberrant secretion of misfolded protein conformations to the extracellular space and exacerbate pathologic protein aggregation into proteotoxic species. Activation of an ER stress-responsive signaling pathway, the Unfolded Protein Response (UPR), restores ER proteostasis through the transcriptional regulation of ER proteostasis pathways. In contrast, the functional role for the UPR in regulating extracellular proteostasis during ER stress is poorly defined. We recently identified ERdj3 as a UPR-regulated secreted chaperone that increases extracellular proteostasis capacity in response to ER stress, revealing a previously-unanticipated direct mechanism by which the UPR impacts extracellular proteostasis. Here, we discuss the functional implications of ERdj3 secretion on extracellular proteostasis maintenance and define the mechanisms by which ERdj3 secretion coordinates intra- and extracellular proteostasis environments during ER stress.
Collapse
Affiliation(s)
- Joseph C Genereux
- a Department of Molecular & Experimental Medicine; Department of Chemical Physiology ; The Scripps Research Institute ; La Jolla , CA USA
| | | |
Collapse
|
28
|
Abstract
Amyloidosis refers to a group of rare but potentially fatal, protein misfolding diseases. The heart is frequently involved in the most common types, that is, immunoglobulin light chain and transthyretin amyloidosis and is the single most important predictor of patient outcomes. A major limitation in improving patient outcomes, in addition to developing novel therapeutics, is the late diagnosis of the disease. Once suspected, an organ for biopsy should be targeted and the amyloid type should be identified by mass spectrometry. An endomyocardial biopsy should be offered if cardiac involvement is in doubt. Echocardiography, MRI and nuclear imaging can provide valuable diagnostic and prognostic information and can secure the diagnosis if amyloid has been identified in an extracardiac tissue.
Collapse
|
29
|
da Costa G, Ribeiro-Silva C, Ribeiro R, Gilberto S, Gomes RA, Ferreira A, Mateus É, Barroso E, Coelho AV, Freire AP, Cordeiro C. Transthyretin Amyloidosis: Chaperone Concentration Changes and Increased Proteolysis in the Pathway to Disease. PLoS One 2015; 10:e0125392. [PMID: 26147092 PMCID: PMC4492746 DOI: 10.1371/journal.pone.0125392] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/24/2015] [Indexed: 11/18/2022] Open
Abstract
Transthyretin amyloidosis is a conformational pathology characterized by the extracellular formation of amyloid deposits and the progressive impairment of the peripheral nervous system. Point mutations in this tetrameric plasma protein decrease its stability and are linked to disease onset and progression. Since non-mutated transthyretin also forms amyloid in systemic senile amyloidosis and some mutation bearers are asymptomatic throughout their lives, non-genetic factors must also be involved in transthyretin amyloidosis. We discovered, using a differential proteomics approach, that extracellular chaperones such as fibrinogen, clusterin, haptoglobin, alpha-1-anti-trypsin and 2-macroglobulin are overrepresented in transthyretin amyloidosis. Our data shows that a complex network of extracellular chaperones are over represented in human plasma and we speculate that they act synergistically to cope with amyloid prone proteins. Proteostasis may thus be as important as point mutations in transthyretin amyloidosis.
Collapse
Affiliation(s)
- Gonçalo da Costa
- Centro de Química e Bioquímica, FCUL, Campo Grande, Lisboa, Portugal
| | | | - Raquel Ribeiro
- Centro de Química e Bioquímica, FCUL, Campo Grande, Lisboa, Portugal
| | - Samuel Gilberto
- Centro de Química e Bioquímica, FCUL, Campo Grande, Lisboa, Portugal
| | - Ricardo A Gomes
- Instituto de Tecnologia Química e Biológica, Av. da República Estação Agronómica Nacional, Oeiras, Portugal
| | - António Ferreira
- Centro de Química e Bioquímica, FCUL, Campo Grande, Lisboa, Portugal
| | - Élia Mateus
- Unidade de Transplantação, Hospital Curry Cabral, Lisboa, Portugal
| | - Eduardo Barroso
- Unidade de Transplantação, Hospital Curry Cabral, Lisboa, Portugal
| | - Ana V Coelho
- Instituto de Tecnologia Química e Biológica, Av. da República Estação Agronómica Nacional, Oeiras, Portugal
| | - Ana Ponces Freire
- Centro de Química e Bioquímica, FCUL, Campo Grande, Lisboa, Portugal
| | - Carlos Cordeiro
- Centro de Química e Bioquímica, FCUL, Campo Grande, Lisboa, Portugal
| |
Collapse
|
30
|
Greene MJ, Klimtchuk ES, Seldin DC, Berk JL, Connors LH. Cooperative stabilization of transthyretin by clusterin and diflunisal. Biochemistry 2014; 54:268-78. [PMID: 25478940 PMCID: PMC4303310 DOI: 10.1021/bi5011249] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
The
circulating protein transthyretin (TTR) can unfold, oligomerize,
and form highly structured amyloid fibrils that are deposited in tissues,
causing organ damage and disease. This pathogenic process is caused
by a heritable TTR point mutation in cases of familial TTR-related
amyloidosis or wild-type TTR in cases of age-associated amyloidosis
(previously called senile systemic amyloidosis). The TTR amyloid cascade
is hypothesized to begin with the dissociation of the TTR native tetrameric
structure into folded but unstable monomeric TTR subunits. Unfolding
of monomeric TTR initiates an oligomerization process leading to aggregation
and fibril formation. Numerous proteostatic mechanisms for regulating
the TTR amyloid cascade exist. Extracellular chaperones provide an
innate defense against misfolded proteins. Clusterin (CLU), a plasma
protein, has the capacity to recognize exposed hydrophobic regions
of misfolded proteins, shielding them from aggregation. We have previously
demonstrated that CLU is associated with the amyloid fibrils in cardiac
tissues from patients with TTR amyloidosis. In this study, we have
used tetrameric and monomeric TTR structural variants to determine
the ability of CLU to inhibit TTR amyloid fibril formation. Using
circular dichroism spectroscopy, we determined that CLU preferentially
stabilizes monomeric TTR and generates increasingly stable conformations
under acid stress. Moreover, studies using surface plasmon resonance
showed a direct interaction of CLU with high-molecular weight TTR
oligomers. The interactions of CLU with monomeric and aggregated TTR
proceed in a cooperative manner in the presence of diflunisal, a small
molecule drug used to stabilize TTR tetramers.
Collapse
Affiliation(s)
- Michael J Greene
- Amyloidosis Center, Boston University School of Medicine , K-507, 715 Albany Street, Boston, Massachusetts 02118, United States
| | | | | | | | | |
Collapse
|
31
|
Sidorova TN, Mace LC, Wells KS, Yermalitskaya LV, Su PF, Shyr Y, Atkinson JB, Fogo AB, Prinsen JK, Byrne JG, Petracek MR, Greelish JP, Hoff SJ, Ball SK, Glabe CG, Brown NJ, Barnett JV, Murray KT. Hypertension is associated with preamyloid oligomers in human atrium: a missing link in atrial pathophysiology? J Am Heart Assoc 2014; 3:e001384. [PMID: 25468655 PMCID: PMC4338732 DOI: 10.1161/jaha.114.001384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Increasing evidence indicates that proteotoxicity plays a pathophysiologic role in experimental and human cardiomyopathy. In organ‐specific amyloidoses, soluble protein oligomers are the primary cytotoxic species in the process of protein aggregation. While isolated atrial amyloidosis can develop with aging, the presence of preamyloid oligomers (PAOs) in atrial tissue has not been previously investigated. Methods and Results Atrial samples were collected during elective cardiac surgery in patients without a history of atrial arrhythmias, congestive heart failure, cardiomyopathy, or amyloidosis. Immunohistochemistry was performed for PAOs using a conformation‐specific antibody, as well as for candidate proteins identified previously in isolated atrial amyloidosis. Using a myocardium‐specific marker, the fraction of myocardium colocalizing with PAOs (PAO burden) was quantified (green/red ratio). Atrial samples were obtained from 92 patients, with a mean age of 61.7±13.8 years. Most patients (62%) were male, 23% had diabetes, 72% had hypertension, and 42% had coronary artery disease. A majority (n=62) underwent aortic valve replacement, with fewer undergoing coronary artery bypass grafting (n=34) or mitral valve replacement/repair (n=24). Immunostaining detected intracellular PAOs in a majority of atrial samples, with a heterogeneous distribution throughout the myocardium. Mean green/red ratio value for the samples was 0.11±0.1 (range 0.03 to 0.77), with a value ≥0.05 in 74 patients. Atrial natriuretic peptide colocalized with PAOs in myocardium, whereas transthyretin was located in the interstitium. Adjusting for multiple covariates, PAO burden was independently associated with the presence of hypertension. Conclusion PAOs are frequently detected in human atrium, where their presence is associated with clinical hypertension.
Collapse
Affiliation(s)
- Tatiana N Sidorova
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| | - Lisa C Mace
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| | - K Sam Wells
- Departments of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN (S.W.)
| | - Liudmila V Yermalitskaya
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| | - Pei-Fang Su
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN (P.F.S., Y.S.)
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN (P.F.S., Y.S.)
| | - James B Atkinson
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN (J.B.A., A.B.F.)
| | - Agnes B Fogo
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN (J.B.A., A.B.F.)
| | - Joseph K Prinsen
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| | - John G Byrne
- Department of Cardiac Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.G.B., M.R.P., J.P.G., S.J.H., S.K.B.)
| | - Michael R Petracek
- Department of Cardiac Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.G.B., M.R.P., J.P.G., S.J.H., S.K.B.)
| | - James P Greelish
- Department of Cardiac Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.G.B., M.R.P., J.P.G., S.J.H., S.K.B.)
| | - Steven J Hoff
- Department of Cardiac Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.G.B., M.R.P., J.P.G., S.J.H., S.K.B.)
| | - Stephen K Ball
- Department of Cardiac Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.G.B., M.R.P., J.P.G., S.J.H., S.K.B.)
| | | | - Nancy J Brown
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| | - Joey V Barnett
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| | - Katherine T Murray
- Department of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (T.N.S., L.C.M., L.V.Y., J.K.P., N.J.B., J.V.B., K.T.M.)
| |
Collapse
|
32
|
Weiss BM, Hebreo J, Cordaro DV, Roschewski MJ, Baker TP, Abbott KC, Olson SW. Increased serum free light chains precede the presentation of immunoglobulin light chain amyloidosis. J Clin Oncol 2014; 32:2699-704. [PMID: 25024082 DOI: 10.1200/jco.2013.50.0892] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Patients with immunoglobulin light chain amyloidosis (AL amyloidosis) generally present with advanced organ dysfunction and have a high risk of early death. We sought to characterize monoclonal immunoglobulin (M-Ig) light chains before clinical presentation of AL amyloidosis. PATIENTS AND METHODS We obtained prediagnostic sera from 20 cases with AL amyloidosis and 20 healthy controls matched for age, sex, race, and age of serum sample from the Department of Defense Serum Repository. Serum protein electrophoresis with immunofixation and serum free light chain (FLC) analysis were performed on all samples. RESULTS An M-Ig was detected in 100% of cases and 0% of controls (P < .001). The M-Ig was present in 100%, 80%, and 42% of cases at less than 4 years, 4 to 11 years, and more than 11 years before diagnosis, respectively. The median FLC differential (FLC-diff) was higher in cases compared with controls at all time periods, less than 4 years (174.8 v 0.3 mg/L; P < .001), 4 to 11 years (65.1 v 2.2 mg/L; P < .001), and more than 11 years (4.5 v 0.4 mg/L; P = .03) before diagnosis. The FLC-diff was greater than 23 mg/L in 85% of cases and 0% of controls (P < .001). The FLC-diff level increased more than 10% per year in 84% of cases compared with 16% of controls (P < .001). CONCLUSION Increase of FLCs, including within the accepted normal range, precedes the development of AL amyloidosis for many years.
Collapse
Affiliation(s)
- Brendan M Weiss
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD.
| | - Joseph Hebreo
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD
| | - Daniel V Cordaro
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD
| | - Mark J Roschewski
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD
| | - Thomas P Baker
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD
| | - Kevin C Abbott
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD
| | - Stephen W Olson
- Brendan M. Weiss, University of Pennsylvania, Philadelphia, PA; Joseph Hebreo, Naval Medical Center San Diego, San Diego, CA; Daniel V. Cordaro, Mark J. Roschewski, Thomas Baker, Kevin C. Abbott, and Stephen W. Olson, Walter Reed National Military Medical Center, Bethesda, MD
| |
Collapse
|
33
|
Brambilla F, Lavatelli F, Di Silvestre D, Valentini V, Palladini G, Merlini G, Mauri P. Shotgun Protein Profile of Human Adipose Tissue and Its Changes in Relation to Systemic Amyloidoses. J Proteome Res 2013; 12:5642-55. [DOI: 10.1021/pr400583h] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
| | - Francesca Lavatelli
- Amyloid
Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Veronica Valentini
- Amyloid
Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giovanni Palladini
- Amyloid
Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department
of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giampaolo Merlini
- Amyloid
Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department
of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Pierluigi Mauri
- Institute for Biomedical Technologies (ITB-CNR), Segrate, Italy
- Institute of Life Sciences, Scuola
Superiore Sant’Anna, Pisa, Italy
| |
Collapse
|
34
|
Esplin BL, Gertz MA. Current Trends in Diagnosis and Management of Cardiac Amyloidosis. Curr Probl Cardiol 2013; 38:53-96. [DOI: 10.1016/j.cpcardiol.2012.11.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
35
|
Franco DA, Truran S, Burciu C, Gutterman DD, Maltagliati A, Weissig V, Hari P, Migrino RQ. Protective role of clusterin in preserving endothelial function in AL amyloidosis. Atherosclerosis 2012; 225:220-3. [PMID: 22981431 DOI: 10.1016/j.atherosclerosis.2012.08.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 08/24/2012] [Accepted: 08/25/2012] [Indexed: 10/27/2022]
Abstract
UNLABELLED Misfolded immunoglobulin light chain proteins (LC) in light chain amyloidosis (AL) are toxic to vascular tissues. We tested the hypothesis that chaperone protein clusterin preserves endothelial function and cell survival during LC exposure. METHODS LC (20 μg/mL) were given to human aortic endothelial cells (EC) for 24-h and clusterin protein/gene expression and secretion were measured. DNA fragmentation was measured with/without recombinant clusterin (Clu, 300 ng/mL). Adipose arterioles (non-AL subjects) were tested for dilator responses to acetylcholine/papaverine at baseline and after 1-h of LC ± Clu. RESULTS LC reduced EC clusterin secretion, protein and gene expression while increasing DNA fragmentation. Clu attenuated LC-induced DNA fragmentation and restored dilator response to acetylcholine (logEC50: control -7.05 ± 0.2, LC + Clu -6.53 ± 0.4, LC -4.28 ± 0.7, p < 0.05 versus control, LC + Clu). CONCLUSIONS LC induced endothelial cell death and dysfunction while reducing clusterin protein/gene expression and secretion. Exogenous clusterin attenuated LC toxicity. This represents a new pathobiologic mechanism and therapeutic target for AL amyloidosis.
Collapse
|
36
|
Roles of Extracellular Chaperones in Amyloidosis. J Mol Biol 2012; 421:499-516. [DOI: 10.1016/j.jmb.2012.01.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 01/24/2023]
|
37
|
Kingsbury JS, Laue TM, Chase SF, Connors LH. Detection of high-molecular-weight amyloid serum protein complexes using biological on-line tracer sedimentation. Anal Biochem 2012; 425:151-6. [PMID: 22465331 PMCID: PMC3354566 DOI: 10.1016/j.ab.2012.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 01/14/2023]
Abstract
The systemic amyloidoses are a rare but deadly class of protein folding disorders with significant unmet diagnostic and therapeutic needs. The current model for symptomatic amyloid progression includes a causative role for soluble toxic aggregates as well as for the fibrillar tissue deposits. Although much research is focused on elucidating the potential mechanism of aggregate toxicity, evidence to support their existence in vivo has been limited. We report the use of a technique we have termed biological on-line tracer sedimentation (BOLTS) to detect abnormal high-molecular-weight complexes (HMWCs) in serum samples from individuals with systemic amyloidosis due to aggregation and deposition of wild-type transthyretin (senile systemic amyloidosis, SSA) or monoclonal immunoglobulin light chain (AL amyloidosis). In this proof-of-concept study, HMWCs were observed in 31 of 77 amyloid samples (40.3%). HMWCs were not detected in any of the 17 nonamyloid control samples subjected to BOLTS analyses. These findings support the existence of potentially toxic amyloid aggregates and suggest that BOLTS may be a useful analytic and diagnostic platform in the study of the amyloidoses or other diseases where abnormal molecular complexes are formed in serum.
Collapse
Affiliation(s)
- Jonathan S. Kingsbury
- Alan and Sandra Gerry Amyloid Research Laboratory in the Amyloid Treatment and Research Program, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Thomas M. Laue
- Department of Biochemistry and Molecular Biology, University of New Hampshire, Durham, New Hampshire 03824, USA
| | - Susan F. Chase
- Department of Biochemistry and Molecular Biology, University of New Hampshire, Durham, New Hampshire 03824, USA
| | - Lawreen H. Connors
- Alan and Sandra Gerry Amyloid Research Laboratory in the Amyloid Treatment and Research Program, Boston University School of Medicine, Boston, Massachusetts 02118, USA
- Department of Biochemistry and Molecular Biology, University of New Hampshire, Durham, New Hampshire 03824, USA
| |
Collapse
|
38
|
|
39
|
Reliable typing of systemic amyloidoses through proteomic analysis of subcutaneous adipose tissue. Blood 2012; 119:1844-7. [DOI: 10.1182/blood-2011-07-365510] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Considering the important advances in treating specific types of systemic amyloidoses, unequivocal typing of amyloid deposits is now essential. Subcutaneous abdominal fat aspiration is the easiest, most common diagnostic procedure. We developed a novel, automated approach, based on Multidimensional Protein Identification Technology, for typing amyloidosis. Fat aspirates were obtained from patients with the most common systemic amyloidoses (ALλ, ALκ, transthyretin, and reactive amyloidosis), with Congo red score more than or equal to 3+, and nonaffected controls. Peptides from extracted and digested proteins were analyzed by Multidimensional Protein Identification Technology. On semiquantitative differential analysis (patients vs controls) of mass spectrometry data, specific proteins up-represented in patients were identified and used as deposit biomarkers. An algorithm was developed to classify patients according to type and abundance of amyloidogenic proteins in samples; in all cases, proteomic characterization was concordant with fibril identification by immunoelectron microscopy and consistent with clinical presentation. Our approach allows reliable amyloid classification using readily available fat aspirates.
Collapse
|
40
|
Dharmarajan K, Maurer MS. Transthyretin cardiac amyloidoses in older North Americans. J Am Geriatr Soc 2012; 60:765-74. [PMID: 22329529 DOI: 10.1111/j.1532-5415.2011.03868.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The amyloidoses are a group of hereditary or acquired disorders caused by the extracellular deposition of insoluble protein fibrils that impair tissue structure and function. All amyloidoses result from protein misfolding, a common mechanism for disorders in older persons, including Alzheimer's disease and Parkinson's disease. Abnormalities in the protein transthyretin (TTR), a serum transporter of thyroxine and retinol, is the most common cause of cardiac amyloidoses in elderly adults. Mutations in TTR can result in familial amyloidotic cardiomyopathy, and wild-type TTR can result in senile cardiac amyloidosis. These underdiagnosed disorders are much more common than previously thought. The resulting restrictive cardiomyopathy can cause congestive heart failure, arrhythmias, and advanced conduction system disease. Although historically difficult to make, the diagnosis of TTR cardiac amyloidosis has become easier in recent years with advances in cardiac imaging and more widespread use of genetic analysis. Although therapy has largely involved supportive medical care, avoidance of potentially toxic agents, and rarely organ transplantation, the near future brings the possibility of targeted pharmacotherapies designed to prevent TTR misfolding and amyloid deposition. Because these disease-modifying agents are designed to prevent disease progression, it has become increasingly important that older persons with TTR amyloidosis be expeditiously identified and considered for enrollment in clinical registries and trials.
Collapse
Affiliation(s)
- Kumar Dharmarajan
- Division of Cardiology, Columbia University Medical Center, New York, New York 10034, USA
| | | |
Collapse
|
41
|
Abstract
There has been much progress in our understanding of transthyretin (TTR)-related amyloidosis including familial amyloidotic polyneuropathy (FAP), senile systemic amyloidosis and its related disorders from many clinical and experimental aspects. FAP is an inherited severe systemic amyloidosis caused by mutated TTR, and characterized by amyloid deposition mainly in the peripheral nervous system and the heart. Liver transplantation is the only available treatment for the disease. FAP is now recognized not to be a rare disease, and to have many variations based on genetical and biochemical variations of TTR. This chapter covers the recent advances in the clinical and pathological aspects of, and therapeutic approaches to FAP, and the trend as to the molecular pathogenesis of TTR.
Collapse
Affiliation(s)
- Takamura Nagasaka
- Department of Neurology, University of Yamanashi, 1110 Shimokato, 409-3898, Chuou-city, Yamanashi, Japan,
| |
Collapse
|
42
|
Clusterin Overexpression and Its Possible Protective Role in Transthyretin Deposition in Familial Amyloidotic Polyneuropathy. J Neuropathol Exp Neurol 2011; 70:1097-106. [DOI: 10.1097/nen.0b013e31823a44f4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
43
|
Azevedo EPC, Pereira HM, Garratt RC, Kelly JW, Foguel D, Palhano FL. Dissecting the structure, thermodynamic stability, and aggregation properties of the A25T transthyretin (A25T-TTR) variant involved in leptomeningeal amyloidosis: identifying protein partners that co-aggregate during A25T-TTR fibrillogenesis in cerebrospinal fluid. Biochemistry 2011; 50:11070-83. [PMID: 22091638 DOI: 10.1021/bi201365r] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Deposition of amorphous aggregates and fibrils of transthyretin (TTR) in leptomeninges and subarachnoid vessels is a characteristic of leptomeningeal amyloidosis (LA), a currently untreatable cerebral angiopathy. Herein, we report the X-ray structure of the A25T homotetramer of TTR, a natural mutant described in a patient with LA. The structure of A25T-TTR is indistinguishable from that of wild-type TTR (wt-TTR), indicating that the difference in amyloidogenicity between A25T-TTR and wt-TTR cannot be ascribed to gross structural differences. Using pressure-induced dissociation of the tetramer, we show that A25T-TTR is 3 kcal/mol less stable than L55P-TTR, the most aggressive mutant of TTR described to date. After incubation for 15 days at 37 °C (pH 7.3), A25T-TTR forms mature amyloid fibrils. To mimic the environment in which TTR aggregates, we investigated aggregation in cerebrospinal fluid (CSF). Unlike L55P-TTR, A25T-TTR rapidly forms amyloid aggregates in CSF that incorporated several protein partners. Utilizing a proteomics methodology, we identified 19 proteins that copurified with A25T-TTR amyloid fibrils. We confirmed the presence of proteins previously identified to be associated with TTR aggregates in biopsies of TTR amyloidosis patients, such as clusterin, apolipoprotein E, and complement proteins. Moreover, we identified novel proteins, such as blood coagulation proteins. Overall, our results revealed the in vitro characterization of TTR aggregation in a biologically relevant environment, opening new avenues of investigation into the molecular mechanisms of LA.
Collapse
Affiliation(s)
- Estefania P C Azevedo
- Instituto de Bioquímica Médica, Programa de Biologia Estrutural, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-590 Brazil
| | | | | | | | | | | |
Collapse
|
44
|
The repertoire of λ light chains causing predominant amyloid heart involvement and identification of a preferentially involved germline gene, IGLV1-44. Blood 2011; 119:144-50. [PMID: 22067386 DOI: 10.1182/blood-2011-05-355784] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monoclonal Ig light chains (LC) can be responsible for pathologic conditions in humans, as in systemic amyloid light amyloidosis. Protean clinical manifestations characterize this disorder with the most varied combination of symptoms generated by different degrees of diverse organ involvement. Kidney and heart are most frequently interested, with major heart involvement as the most relevant prognostic factor. The identification of the underlying mechanism involved in organ targeting is of major relevance for the pathobiology of this disorder. To this aim, we characterized the repertoire of variable region germline genes of λ LC preferentially targeting the heart and compared it with the repertoire of LC that do not in a case-control study. We found that the repertoires were highly restricted, showing preferential use of the same few germline genes but with a different frequency pattern. A single gene, IGVL1-44, was found associated with a 5-fold increase in the odds of dominant heart involvement (after adjusting for confounders in a multivariable logistic model). These results support an involvement of LC genetics in the determination of organ targeting. Study of the characteristics of IGVL1-44-LC with, and of the minority without, heart involvement might lead to identification of LC/tissue interactions.
Collapse
|
45
|
Guan J, Mishra S, Falk RH, Liao R. Current perspectives on cardiac amyloidosis. Am J Physiol Heart Circ Physiol 2011; 302:H544-52. [PMID: 22058156 DOI: 10.1152/ajpheart.00815.2011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amyloidosis represents a group of diseases in which proteins undergo misfolding to form insoluble fibrils with subsequent tissue deposition. While almost all deposited amyloid fibers share a common nonbranched morphology, the affected end organs, clinical presentation, treatment strategies, and prognosis vary greatly among this group of diseases and are largely dependent on the specific amyloid precursor protein. To date, at least 27 precursor proteins have been identified to result in either local tissue or systemic amyloidosis, with nine of them manifesting in cardiac deposition and resulting in a syndrome termed "cardiac amyloidosis" or "amyloid cardiomyopathy." Although cardiac amyloidosis has been traditionally considered to be a rare disorder, as clinical appreciation and understanding continues to grow, so too has the prevalence, suggesting that this disease may be greatly underdiagnosed. The most common form of cardiac amyloidosis is associated with circulating amyloidogenic monoclonal immunoglobulin light chain proteins. Other major cardiac amyloidoses result from a misfolding of products of mutated or wild-type transthyretin protein. While the various cardiac amyloidoses share a common functional consequence, namely, an infiltrative cardiomyopathy with restrictive pathophysiology leading to progressive heart failure, the underlying pathophysiology and clinical syndrome varies with each precursor protein. Herein, we aim to provide an up-to-date overview of cardiac amyloidosis from nomenclature to molecular mechanisms and treatment options, with a particular focus on amyloidogenic immunoglobulin light chain protein cardiac amyloidosis.
Collapse
Affiliation(s)
- Jian Guan
- Cardiac Muscle Research Lab., 77 Ave. Louis Pasteur, NRB 431, Boston, MA 02115, USA
| | | | | | | |
Collapse
|