1
|
Lederberg OL, Yan NL, Sanchez J, Ren W, Ash C, Wilkens SJ, Qiu H, Qin B, Grant VH, Jackman AB, Stanfield RL, Wilson IA, Petrassi HM, Rhoades D, Kelly JW. Discovery of Potent and Selective Pyridone-Based Small Molecule Kinetic Stabilizers of Amyloidogenic Immunoglobulin Light Chains. J Med Chem 2024; 67:21070-21105. [PMID: 39626211 DOI: 10.1021/acs.jmedchem.4c01773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Kinetic stabilization of amyloidogenic immunoglobulin light chains (LCs) through small molecule binding may become the first treatment for the proteinopathy component of light chain amyloidosis (AL). Kinetic stabilizers selectively bind to the native state over the misfolding transition state, slowing denaturation. Prior λ full-length LC dimer (FL LC2) kinetic stabilizers exhibited considerable plasma protein binding. We hypothesized that the coumarin "aromatic core" of the stabilizers was responsible for the undesirable plasma protein binding. Here, we describe structure-activity relationship (SAR) data initially focused on replacing the coumarin aromatic core. 2-pyridones proved suitable replacements. We subsequently optimized the "anchor substructure" in the context of 2-pyridones, resulting in potent λ FL LC2 kinetic stabilizers exhibiting reduced plasma protein binding. The 3-methyl- or 3-ethyl-3-phenylpyrrolidine-2-pyridone scaffold stabilized multiple AL patient-derived λ FL LC2s in human plasma. This, coupled with X-ray crystallographic data, indicates that 3-alkyl-3-phenylpyrrolidine-2-pyridone-based stabilizers are promising candidates for treating the proteinopathy component of AL.
Collapse
Affiliation(s)
- Oren L Lederberg
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Nicholas L Yan
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Julian Sanchez
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Wen Ren
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Carl Ash
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Steven J Wilkens
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Huang Qiu
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Bo Qin
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Virginia H Grant
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Alex B Jackman
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road., La Jolla, California 92037, United States
| | - H Michael Petrassi
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Derek Rhoades
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road., La Jolla, California 92037, United States
| |
Collapse
|
2
|
Palladini G, Liedtke M, Zago W, Dolan P, Kinney GG, Gertz MA. The mechanism of action, pharmacological characteristics, and clinical utility of the amyloid depleter birtamimab for the potential treatment of AL amyloidosis. Leuk Lymphoma 2024; 65:1068-1078. [PMID: 38600883 DOI: 10.1080/10428194.2024.2337803] [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: 12/04/2023] [Accepted: 03/27/2024] [Indexed: 04/12/2024]
Abstract
Amyloid light chain (AL) amyloidosis is a progressive plasma cell disorder caused by amyloid deposition resulting in organ damage and failure. Current standard-of-care treatments target clonal plasma cells, the source of misfolded light chains (amyloid precursors), yet only half of patients with advanced disease survive ≥6 months. The amyloid depleter birtamimab is an investigational humanized monoclonal antibody that binds misfolded κ and λ light chains with high specificity and was designed to neutralize soluble toxic light chain aggregates and promote phagocytic clearance of deposited amyloid. Post hoc analyses from the Phase 3 VITAL trial suggested birtamimab plus standard of care confers a survival benefit in patients with advanced (Mayo Stage IV) AL amyloidosis. AFFIRM-AL (NCT04973137), a Phase 3 confirmatory trial of birtamimab plus standard of care in patients with Mayo Stage IV AL amyloidosis, is ongoing. This review summarizes birtamimab's mechanism of action, attributes, and potential clinical utility.
Collapse
Affiliation(s)
- Giovanni Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Fondazione IRCCS, Policlinico San Matteo, Pavia, Italy
| | | | | | - Phil Dolan
- Prothena Biosciences Inc, Brisbane, CA, USA
| | | | - Morie A Gertz
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Desantis F, Miotto M, Milanetti E, Ruocco G, Di Rienzo L. Computational evidences of a misfolding event in an aggregation-prone light chain preceding the formation of the non-native pathogenic dimer. Proteins 2024; 92:797-807. [PMID: 38314653 DOI: 10.1002/prot.26672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/09/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Antibody light chain amyloidosis is a disorder in which protein aggregates, mainly composed of immunoglobulin light chains, deposit in diverse tissues impairing the correct functioning of organs. Interestingly, due to the high susceptibility of antibodies to mutations, AL amyloidosis appears to be strongly patient-specific. Indeed, every patient will display their own mutations that will make the proteins involved prone to aggregation thus hindering the study of this disease on a wide scale. In this framework, determining the molecular mechanisms that drive the aggregation could pave the way to the development of patient-specific therapeutics. Here, we focus on a particular patient-derived light chain, which has been experimentally characterized. We investigated the early phases of the aggregation pathway through extensive full-atom molecular dynamics simulations, highlighting a structural rearrangement and the exposure of two hydrophobic regions in the aggregation-prone species. Next, we moved to consider the pathological dimerization process through docking and molecular dynamics simulations, proposing a dimeric structure as a candidate pathological first assembly. Overall, our results shed light on the first phases of the aggregation pathway for a light chain at an atomic level detail, offering new structural insights into the corresponding aggregation process.
Collapse
Affiliation(s)
- Fausta Desantis
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia, Genova, Italy
- Istituto Italiano di Tecnologia (IIT), Center for Life Nano & Neuro Science, Roma, Italy
| | - Mattia Miotto
- Istituto Italiano di Tecnologia (IIT), Center for Life Nano & Neuro Science, Roma, Italy
| | - Edoardo Milanetti
- Istituto Italiano di Tecnologia (IIT), Center for Life Nano & Neuro Science, Roma, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- Istituto Italiano di Tecnologia (IIT), Center for Life Nano & Neuro Science, Roma, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Di Rienzo
- Istituto Italiano di Tecnologia (IIT), Center for Life Nano & Neuro Science, Roma, Italy
| |
Collapse
|
4
|
Karimi-Farsijani S, Pfeiffer PB, Banerjee S, Baur J, Kuhn L, Kupfer N, Hegenbart U, Schönland SO, Wiese S, Haupt C, Schmidt M, Fändrich M. Light chain mutations contribute to defining the fibril morphology in systemic AL amyloidosis. Nat Commun 2024; 15:5121. [PMID: 38879609 PMCID: PMC11180120 DOI: 10.1038/s41467-024-49520-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 06/06/2024] [Indexed: 06/19/2024] Open
Abstract
Systemic AL amyloidosis is one of the most frequently diagnosed forms of systemic amyloidosis. It arises from mutational changes in immunoglobulin light chains. To explore whether these mutations may affect the structure of the formed fibrils, we determine and compare the fibril structures from several patients with cardiac AL amyloidosis. All patients are affected by light chains that contain an IGLV3-19 gene segment, and the deposited fibrils differ by the mutations within this common germ line background. Using cryo-electron microscopy, we here find different fibril structures in each patient. These data establish that the mutations of amyloidogenic light chains contribute to defining the fibril architecture and hence the structure of the pathogenic agent.
Collapse
Affiliation(s)
| | | | | | - Julian Baur
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Lukas Kuhn
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Niklas Kupfer
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Ute Hegenbart
- Medicinal Department V, Amyloidosis Centre, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan O Schönland
- Medicinal Department V, Amyloidosis Centre, Heidelberg University Hospital, Heidelberg, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany
| | - Christian Haupt
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | | | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| |
Collapse
|
5
|
Lavatelli F, Natalello A, Marchese L, Ami D, Corazza A, Raimondi S, Mimmi MC, Malinverni S, Mangione PP, Palmer MT, Lampis A, Concardi M, Verona G, Canetti D, Arbustini E, Bellotti V, Giorgetti S. Truncation of the constant domain drives amyloid formation by immunoglobulin light chains. J Biol Chem 2024; 300:107174. [PMID: 38499153 PMCID: PMC11016911 DOI: 10.1016/j.jbc.2024.107174] [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: 12/08/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
AL amyloidosis is a life-threatening disease caused by deposition of immunoglobulin light chains. While the mechanisms underlying light chains amyloidogenesis in vivo remain unclear, several studies have highlighted the role that tissue environment and structural amyloidogenicity of individual light chains have in the disease pathogenesis. AL natural deposits contain both full-length light chains and fragments encompassing the variable domain (VL) as well as different length segments of the constant region (CL), thus highlighting the relevance that proteolysis may have in the fibrillogenesis pathway. Here, we investigate the role of major truncated species of the disease-associated AL55 light chain that were previously identified in natural deposits. Specifically, we study structure, molecular dynamics, thermal stability, and capacity to form fibrils of a fragment containing both the VL and part of the CL (133-AL55), in comparison with the full-length protein and its variable domain alone, under shear stress and physiological conditions. Whereas the full-length light chain forms exclusively amorphous aggregates, both fragments generate fibrils, although, with different kinetics, aggregate structure, and interplay with the unfragmented protein. More specifically, the VL-CL 133-AL55 fragment entirely converts into amyloid fibrils microscopically and spectroscopically similar to their ex vivo counterpart and increases the amorphous aggregation of full-length AL55. Overall, our data support the idea that light chain structure and proteolysis are both relevant for amyloidogenesis in vivo and provide a novel biocompatible model of light chain fibrillogenesis suitable for future mechanistic studies.
Collapse
Affiliation(s)
- Francesca Lavatelli
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.
| | - Loredana Marchese
- Pathology Unit, Fondazione IRCSS Policlinico San Matteo, Pavia, Italy
| | - Diletta Ami
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Alessandra Corazza
- Department of Medicine (DAME), University of Udine, Udine, Italy; Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Sara Raimondi
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
| | - Maria Chiara Mimmi
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Silvia Malinverni
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
| | - P Patrizia Mangione
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Manel Terrones Palmer
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Alessio Lampis
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
| | - Monica Concardi
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Guglielmo Verona
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Centre for Amyloidosis, Division of Medicine, University College London, London, UK
| | - Diana Canetti
- Centre for Amyloidosis, Division of Medicine, University College London, London, UK
| | - Eloisa Arbustini
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Vittorio Bellotti
- Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sofia Giorgetti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
6
|
Broggini L, Barzago MM, Speranzini V, Schulte T, Sonzini F, Giono M, Romeo M, Milani P, Caminito S, Mazzini G, Rognoni P, Merlini G, Pappone C, Anastasia L, Nuvolone M, Palladini G, Diomede L, Ricagno S. Nanobodies counteract the toxicity of an amyloidogenic light chain by stabilizing a partially open dimeric conformation. J Mol Biol 2023; 435:168320. [PMID: 37865287 DOI: 10.1016/j.jmb.2023.168320] [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: 06/22/2023] [Revised: 09/18/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Light chain amyloidosis (AL) is a systemic disease where fibrillar deposition of misfolded immunoglobulin light chains (LCs) severely affects organ function and results in poor prognosis for patients, especially when heart involvement is severe. Particularly relevant in this context is the cardiotoxicity exerted by still uncharacterized soluble LC species. Here, with the final goal of identifying alternative therapeutic strategies to tackle AL amyloidosis, we produced five llama-derived nanobodies (Nbs) specific against H3, a well-characterized amyloidogenic and cardiotoxic LC from an AL patient with severe cardiac involvement. We found that Nbs are specific and potent agents capable of abolishing H3 soluble toxicity in C. elegans in vivo model. Structural characterization of H3-Nb complexes revealed that the protective effect of Nbs is related to their ability to bind to the H3 VL domain and stabilise an unexpected partially open LC dimer in which the two VL domains no longer interact with each other. Thus, while identifying potent inhibitors of LC soluble toxicity, we also describe the first non-native structure of an amyloidogenic LC that may represent a crucial step in toxicity and aggregation mechanisms.
Collapse
Affiliation(s)
- Luca Broggini
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Piazza Malan 2, 20097 San Donato Milanese, Italy
| | - Maria Monica Barzago
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, Milano 20156, Italy
| | | | - Tim Schulte
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Piazza Malan 2, 20097 San Donato Milanese, Italy
| | - Federica Sonzini
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Piazza Malan 2, 20097 San Donato Milanese, Italy; Department of Biosciences, Università degli Studi di Milano, Milan 20133, Italy
| | - Matteo Giono
- Department of Biosciences, Università degli Studi di Milano, Milan 20133, Italy
| | - Margherita Romeo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, Milano 20156, Italy
| | - Paolo Milani
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Serena Caminito
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Giulia Mazzini
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Paola Rognoni
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Giampaolo Merlini
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Carlo Pappone
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Piazza Malan 2, 20097 San Donato Milanese, Italy; Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, San Donato, Milan 20097, Italy; Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Luigi Anastasia
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Piazza Malan 2, 20097 San Donato Milanese, Italy; Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Mario Nuvolone
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Giovanni Palladini
- Amyloidosis Treatment and Research Center, Fondazione IRCCS Policlinico San Matteo, Università Degli Studi di Pavia, Pavia 27100, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, Milano 20156, Italy
| | - Stefano Ricagno
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Piazza Malan 2, 20097 San Donato Milanese, Italy; Department of Biosciences, Università degli Studi di Milano, Milan 20133, Italy.
| |
Collapse
|
7
|
Klimtchuk ES, Peterle D, Bullitt EA, Connors LH, Engen JR, Gursky O. Role of complementarity-determining regions 1 and 3 in pathologic amyloid formation by human immunoglobulin κ1 light chains. Amyloid 2023; 30:364-378. [PMID: 37216473 PMCID: PMC10663386 DOI: 10.1080/13506129.2023.2212397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Immunoglobulin light chain (LC) amyloidosis is a life-threatening disease complicated by vast numbers of patient-specific mutations. We explored 14 patient-derived and engineered proteins related to κ1-family germline genes IGKVLD-33*01 and IGKVLD-39*01. METHODS Hydrogen-deuterium exchange mass spectrometry analysis of conformational dynamics in recombinant LCs and their fragments was integrated with studies of thermal stability, proteolytic susceptibility, amyloid formation and amyloidogenic sequence propensity. The results were mapped on the structures of native and fibrillary proteins. RESULTS Proteins from two κ1 subfamilies showed unexpected differences. Compared to their germline counterparts, amyloid LC related to IGKVLD-33*01 was less stable and formed amyloid faster, whereas amyloid LC related to IGKVLD-39*01 had similar stability and formed amyloid slower, suggesting different major factors influencing amyloidogenesis. In 33*01-related amyloid LC, these factors involved destabilization of the native structure and probable stabilization of amyloid. The atypical behavior of 39*01-related amyloid LC stemmed from increased dynamics/exposure of amyloidogenic segments in βC'V and βEV that could initiate aggregation and decreased dynamics/exposure near the Cys23-Cys88 disulfide. CONCLUSIONS The results suggest distinct amyloidogenic pathways for closely related LCs and point to the complementarity-defining regions CDR1 and CDR3, linked via the conserved internal disulfide, as key factors in amyloid formation.
Collapse
Affiliation(s)
- Elena S. Klimtchuk
- Amyloidosis Center, Boston University Chobanian and Avedisian School of Medicine, Boston MA 02118, United States
| | - Daniele Peterle
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States
| | - Esther A. Bullitt
- Department of Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, W302, 700 Albany Street, Boston, MA, 02118, United States
| | - Lawreen H. Connors
- Amyloidosis Center, Boston University Chobanian and Avedisian School of Medicine, Boston MA 02118, United States
| | - John R. Engen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States
| | - Olga Gursky
- Amyloidosis Center, Boston University Chobanian and Avedisian School of Medicine, Boston MA 02118, United States
- Department of Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, W302, 700 Albany Street, Boston, MA, 02118, United States
| |
Collapse
|
8
|
Pradhan T, Sarkar R, Meighen-Berger KM, Feige MJ, Zacharias M, Reif B. Mechanistic insights into the aggregation pathway of the patient-derived immunoglobulin light chain variable domain protein FOR005. Nat Commun 2023; 14:3755. [PMID: 37353525 PMCID: PMC10290123 DOI: 10.1038/s41467-023-39280-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 06/05/2023] [Indexed: 06/25/2023] Open
Abstract
Systemic antibody light chain (AL) amyloidosis is characterized by deposition of amyloid fibrils. Prior to fibril formation, soluble oligomeric AL protein has a direct cytotoxic effect on cardiomyocytes. We focus on the patient derived λ-III AL variable domain FOR005 which is mutated at five positions with respect to the closest germline protein. Using solution-state NMR spectroscopy, we follow the individual steps involved in protein misfolding from the native to the amyloid fibril state. Unfavorable mutations in the complementary determining regions introduce a strain in the native protein structure which yields partial unfolding. Driven by electrostatic interactions, the protein converts into a high molecular weight, oligomeric, molten globule. The high local concentration of aggregation prone regions in the oligomer finally catalyzes the conversion into fibrils. The topology is determined by balanced electrostatic interactions in the fibril core implying a 180° rotational switch of the beta-sheets around the conserved disulfide bond.
Collapse
Affiliation(s)
- Tejaswini Pradhan
- Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University Munich, Lichtenbergstr. 4, 85747, Garching, Germany
- Institute of Structural Biology (STB), Helmholtz-Zentrum München (HMGU), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Riddhiman Sarkar
- Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University Munich, Lichtenbergstr. 4, 85747, Garching, Germany
- Institute of Structural Biology (STB), Helmholtz-Zentrum München (HMGU), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Kevin M Meighen-Berger
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Matthias J Feige
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Martin Zacharias
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Bernd Reif
- Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University Munich, Lichtenbergstr. 4, 85747, Garching, Germany.
- Institute of Structural Biology (STB), Helmholtz-Zentrum München (HMGU), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
| |
Collapse
|
9
|
Baur J, Berghaus N, Schreiner S, Hegenbart U, Schönland SO, Wiese S, Huhn S, Haupt C. Identification of AL proteins from 10 λ-AL amyloidosis patients by mass spectrometry extracted from abdominal fat and heart tissue. Amyloid 2023; 30:27-37. [PMID: 35792725 DOI: 10.1080/13506129.2022.2095618] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Systemic AL amyloidosis arises from the misfolding of patient-specific immunoglobulin light chains (LCs). Potential drivers of LC amyloid formation are mutational changes and post-translational modifications (PTMs). However, little information is available on the exact primary structure of the AL proteins and their precursor LCs. OBJECTIVE We analyse the exact primary structure of AL proteins extracted from 10 λ AL amyloidosis patients and their corresponding precursor LCs. MATERIALS AND METHODS By cDNA sequencing of the precursor LC genes in combination with mass spectrometry of the AL proteins, the exact primary structure and PTMs were determined. This information was used to analyse their biochemical properties. RESULTS All AL proteins comprise the VL and a small part of the CL with a common C-terminal truncation region. While all AL proteins retain the conserved native disulphide bond of the VL, we found no evidence for presence of other common PTMs. The analysis of the biochemical properties revealed that the isoelectric point of the VL is significantly increased due to introduced mutations. CONCLUSION Our data imply that mutational changes influence the surface charge properties of the VL and that common proteolytic processes are involved in the generation of the cleavage sites of AL proteins.
Collapse
Affiliation(s)
- Julian Baur
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Natalie Berghaus
- Medical Department V, Amyloidosis Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Sarah Schreiner
- Medical Department V, Amyloidosis Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Ute Hegenbart
- Medical Department V, Amyloidosis Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan O Schönland
- Medical Department V, Amyloidosis Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Medical Faculty, Ulm University, Ulm, Germany
| | - Stefanie Huhn
- Medical Department V, Section of Multiple Myeloma, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Haupt
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| |
Collapse
|
10
|
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: 1] [Impact Index Per Article: 0.5] [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
|
11
|
Rottenaicher GJ, Absmeier RM, Meier L, Zacharias M, Buchner J. A constant domain mutation in a patient-derived antibody light chain reveals principles of AL amyloidosis. Commun Biol 2023; 6:209. [PMID: 36823438 PMCID: PMC9950467 DOI: 10.1038/s42003-023-04574-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Light chain (AL) amyloidosis is a debilitating disease in which mutant antibody light chains (LC), secreted by aberrant plasma cell clones, misfold and form insoluble fibrils, which can be deposited in various organs. In the majority of cases, the fibrillar deposits consist of LC variable domains (VL) containing destabilizing mutations compared to their germline counterparts. This is also true for the patient LC FOR005. However, this pathogenic LC sequence contains an additional mutation in the constant domain (CL). The mechanistic impact of CL mutations is not yet understood in the context of AL amyloidosis. Our analysis reveals that the FOR005 CL mutation influences the amyloid pathway in specific ways: (1) folding and stability of the patient CL domain are strongly impaired; (2) the mutation disrupts the LC dimer interface and weakens dimerization; (3) the CL mutation promotes proteolytic cleavage of the LC monomers resulting in an isolated, amyloidogenic VL domain while dimeric LCs are not cleaved. The enhanced proteolysis rates and the inability of full-length LCs to form amyloid fibrils even in the presence of a destabilized CL domain support a model for AL amyloidosis in which the CL domain plays a protective role and in which proteolytic cleavage precedes amyloid formation.
Collapse
Affiliation(s)
- Georg J Rottenaicher
- Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
- Department of Biosciences, TUM School of Natural Sciences, Technical University Munich, Boltzmannstr. 10, 85748, Garching, Germany
| | - Ramona M Absmeier
- Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
- Department of Biosciences, TUM School of Natural Sciences, Technical University Munich, Boltzmannstr. 10, 85748, Garching, Germany
| | - Laura Meier
- Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
- Department of Biosciences, TUM School of Natural Sciences, Technical University Munich, Boltzmannstr. 10, 85748, Garching, Germany
| | - Martin Zacharias
- Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
- Department of Biosciences, TUM School of Natural Sciences, Technical University Munich, Boltzmannstr. 10, 85748, Garching, Germany
| | - Johannes Buchner
- Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany.
- Department of Biosciences, TUM School of Natural Sciences, Technical University Munich, Boltzmannstr. 10, 85748, Garching, Germany.
| |
Collapse
|
12
|
Klimtchuk ES, Peterle D, Bullitt EA, Connors LH, Engen JR, Gursky O. Role of Complementarity-Determining Regions 1 and 3 in Pathologic Amyloid Formation by Human Immunoglobulin κ1 Light Chains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526662. [PMID: 36778378 PMCID: PMC9915687 DOI: 10.1101/2023.02.01.526662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunoglobulin light chain (LC) amyloidosis is a life-threatening disease whose understanding and treatment is complicated by vast numbers of patient-specific mutations. To address molecular origins of the disease, we explored 14 patient-derived and engineered proteins related to κ1-family germline genes IGKVLD-33*01 and IGKVLD-39*01. Hydrogen-deuterium exchange mass spectrometry analysis of local conformational dynamics in full-length recombinant LCs and their fragments was integrated with studies of thermal stability, proteolytic susceptibility, amyloid formation, and amyloidogenic sequence propensities using spectroscopic, electron microscopic and bioinformatics tools. The results were mapped on the atomic structures of native and fibrillary proteins. Proteins from two κ1 subfamilies showed unexpected differences. Compared to their germline counterparts, amyloid LC related to IGKVLD-33*01 was less stable and formed amyloid faster, whereas amyloid LC related to IGKVLD-39*01 had similar stability and formed amyloid slower. These and other differences suggest different major factors influencing amyloid formation. In 33*01-related amyloid LC, these factors involved mutation-induced destabilization of the native structure and probable stabilization of amyloid. The atypical behaviour of 39*01-related amyloid LC tracked back to increased dynamics/exposure of amyloidogenic segments in βC' V and βE V that could initiate aggregation, combined with decreased dynamics/exposure near the Cys23-Cys88 disulfide whose rearrangement is rate-limiting to amyloidogenesis. The results suggest distinct amyloidogenic pathways for closely related LCs and point to the antigen-binding, complementarity-determining regions CDR1 and CDR3, which are linked via the conserved internal disulfide, as key factors in amyloid formation by various LCs.
Collapse
|
13
|
An N-glycosylation hotspot in immunoglobulin κ light chains is associated with AL amyloidosis. Leukemia 2022; 36:2076-2085. [PMID: 35610346 DOI: 10.1038/s41375-022-01599-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Immunoglobulin light chain (AL) amyloidosis is caused by a small, minimally proliferating B-cell/plasma-cell clone secreting a patient-unique, aggregation-prone, toxic light chain (LC). The pathogenicity of LCs is encrypted in their sequence, yet molecular determinants of amyloidogenesis are poorly understood. Higher rates of N-glycosylation among clonal κ LCs from patients with AL amyloidosis compared to other monoclonal gammopathies indicate that this post-translational modification is associated with a higher risk of developing AL amyloidosis. Here, we exploited LC sequence information from previously published amyloidogenic and control clonal LCs and from a series of 220 patients with AL amyloidosis or multiple myeloma followed at our Institutions to define sequence and spatial features of N-glycosylation, combining bioinformatics, biochemical, proteomics, structural and genetic analyses. We found peculiar sequence and spatial pattern of N-glycosylation in amyloidogenic κ LCs, with most of the N-glycosylation sites laying in the framework region 3, particularly within the E strand, and consisting mainly of the NFT sequon, setting them apart with respect to non-amyloidogenic clonal LCs. Our data further support a potential role of N-glycosylation in determining the pathogenic behavior of a subset of amyloidogenic LCs and may help refine current N-glycosylation-based prognostic assessments for patients with monoclonal gammopathies.
Collapse
|
14
|
Yan NL, Nair R, Chu A, Wilson IA, Johnson KA, Morgan GJ, Kelly JW. Amyloidogenic immunoglobulin light chain kinetic stabilizers comprising a simple urea linker module reveal a novel binding sub-site. Bioorg Med Chem Lett 2022; 60:128571. [PMID: 35065233 PMCID: PMC8857066 DOI: 10.1016/j.bmcl.2022.128571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/29/2021] [Accepted: 01/15/2022] [Indexed: 11/29/2022]
Abstract
In immunoglobulin light chain (LC) amyloidosis, the misfolding, or misfolding and misassembly of LC a protein or fragments thereof resulting from aberrant endoproteolysis, causes organ damage to patients. A small molecule "kinetic stabilizer" drug could slow or stop these processes and improve prognosis. We previously identified coumarin-based kinetic stabilizers of LCs that can be divided into four components, including a "linker module" and "distal substructure". Our prior studies focused on characterizing carbamate, hydantoin, and spirocyclic urea linker modules, which bind in a solvent-exposed site at the VL-VL domain interface of the LC dimer. Here, we report structure-activity relationship data on 7-diethylamino coumarin-based kinetic stabilizers. This substructure occupies the previously characterized "anchor cavity" and the "aromatic slit". The potencies of amide and urea linker modules terminating in a variety of distal substructures attached at the 3-position of this coumarin ring were assessed. Surprisingly, crystallographic data on a 7-diethylamino coumarin-based kinetic stabilizer reveals that the urea linker module and distal substructure attached at the 3-position bind a solvent-exposed region of the full-length LC dimer distinct from previously characterized sites. Our results further elaborate the small-molecule binding surface of LCs that could be occupied by potent and selective LC kinetic stabilizers.
Collapse
Affiliation(s)
- Nicholas L Yan
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Reji Nair
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alan Chu
- California Institute for Biomedical Research, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kristen A Johnson
- California Institute for Biomedical Research, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Gareth J Morgan
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; The Amyloidosis Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
15
|
Stelmach-Gołdyś A, Zaborek-Łyczba M, Łyczba J, Garus B, Pasiarski M, Mertowska P, Małkowska P, Hrynkiewicz R, Niedźwiedzka-Rystwej P, Grywalska E. Physiology, Diagnosis and Treatment of Cardiac Light Chain Amyloidosis. J Clin Med 2022; 11:jcm11040911. [PMID: 35207184 PMCID: PMC8880759 DOI: 10.3390/jcm11040911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
AL (light-chain) amyloidosis is a systemic disease in which amyloid fibers are formed from kappa or lambda immunoglobulin light chains, or fragments thereof, produced by a neoplastic clone of plasmocytes. The produced protein is deposited in tissues and organs in the form of extracellular deposits, which leads to impairment of their functions and, consequently, to death. Despite the development of research on pathogenesis and therapy, the mortality rate of patients with late diagnosed amyloidosis is 30%. The diagnosis is delayed due to the complex clinical picture and the slow progression of the disease. This is the type of amyloidosis that most often contributes to cardiac lesions and causes cardiac amyloidosis (CA). Early diagnosis and correct identification of the type of amyloid plays a crucial role in the planning and effectiveness of therapy. In addition to standard histological studies based on Congo red staining, diagnostics are enriched by tests to determine the degree of cardiac involvement. In this paper, we discuss current diagnostic methods used in cardiac light chain amyloidosis and the latest therapies that contribute to an improved patient prognosis.
Collapse
Affiliation(s)
- Agnieszka Stelmach-Gołdyś
- Department of Immunology, Faculty of Health Sciences, Jan Kochanowski University, 25-317 Kielce, Poland; (A.S.-G.); (B.G.); (M.P.)
- Department of Hematology, Holy Cross Cancer Centre, 25-734 Kielce, Poland
| | - Monika Zaborek-Łyczba
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (M.Z.-Ł.); (J.Ł.); (P.M.); (E.G.)
| | - Jakub Łyczba
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (M.Z.-Ł.); (J.Ł.); (P.M.); (E.G.)
| | - Bartosz Garus
- Department of Immunology, Faculty of Health Sciences, Jan Kochanowski University, 25-317 Kielce, Poland; (A.S.-G.); (B.G.); (M.P.)
- Department of Hematology, Holy Cross Cancer Centre, 25-734 Kielce, Poland
| | - Marcin Pasiarski
- Department of Immunology, Faculty of Health Sciences, Jan Kochanowski University, 25-317 Kielce, Poland; (A.S.-G.); (B.G.); (M.P.)
- Department of Hematology, Holy Cross Cancer Centre, 25-734 Kielce, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (M.Z.-Ł.); (J.Ł.); (P.M.); (E.G.)
| | - Paulina Małkowska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (R.H.)
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (R.H.)
| | | | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (M.Z.-Ł.); (J.Ł.); (P.M.); (E.G.)
| |
Collapse
|
16
|
Lewkowicz E, Gursky O. Dynamic protein structures in normal function and pathologic misfolding in systemic amyloidosis. Biophys Chem 2022; 280:106699. [PMID: 34773861 PMCID: PMC9416430 DOI: 10.1016/j.bpc.2021.106699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 02/08/2023]
Abstract
Dynamic and disordered regions in native proteins are often critical for their function, particularly in ligand binding and signaling. In certain proteins, however, such regions can contribute to misfolding and pathologic deposition as amyloid fibrils in vivo. For example, dynamic and disordered regions can promote amyloid formation by destabilizing the native structure, by directly triggering the aggregation, by promoting protein condensation, or by acting as sites of early proteolytic cleavage that favor a release of aggregation-prone fragments or facilitate fibril maturation. At the same time, enhanced dynamics in the native protein state accelerates proteolytic degradation that counteracts amyloid accumulation in vivo. Therefore, the functional need for dynamic protein regions must be balanced against their inherently labile nature. How exactly this balance is achieved and how is it shifted upon amyloidogenic mutations or post-translational modifications? To illustrate possible scenarios, here we review the beneficial and pathologic roles of dynamic and disordered regions in the native states of three families of human plasma proteins that form amyloid precursors in systemic amyloidoses: immunoglobulin light chain, apolipoproteins, and serum amyloid A. Analysis of structure, stability and local dynamics of these diverse proteins and their amyloidogenic variants exemplifies how disordered/dynamic regions can provide a functional advantage as well as an Achilles heel in pathologic amyloid formation.
Collapse
|
17
|
A Conservative Point Mutation in a Dynamic Antigen-binding Loop of Human Immunoglobulin λ6 Light Chain Promotes Pathologic Amyloid Formation. J Mol Biol 2021; 433:167310. [PMID: 34678302 DOI: 10.1016/j.jmb.2021.167310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/07/2023]
Abstract
Immunoglobulin light chain (LC) amyloidosis (AL) is a life-threatening human disease wherein free mono-clonal LCs deposit in vital organs. To determine what makes some LCs amyloidogenic, we explored patient-based amyloidogenic and non-amyloidogenic recombinant LCs from the λ6 subtype prevalent in AL. Hydrogen-deuterium exchange mass spectrometry, structural stability, proteolysis, and amyloid growth studies revealed that the antigen-binding CDR1 loop is the least protected part in the variable domain of λ6 LC, particularly in the AL variant. N32T substitution in CRD1 is identified as a driver of amyloid formation. Substitution N32T increased the amyloidogenic propensity of CDR1 loop, decreased its protection in the native structure, and accelerated amyloid growth in the context of other AL substitutions. The destabilizing effects of N32T propagated across the molecule increasing its dynamics in regions ∼30 Å away from the substitution site. Such striking long-range effects of a conservative point substitution in a dynamic surface loop may be relevant to Ig function. Comparison of patient-derived and engineered proteins showed that N32T interactions with other substitution sites must contribute to amyloidosis. The results suggest that CDR1 is critical in amyloid formation by other λ6 LCs.
Collapse
|
18
|
Radamaker L, Karimi-Farsijani S, Andreotti G, Baur J, Neumann M, Schreiner S, Berghaus N, Motika R, Haupt C, Walther P, Schmidt V, Huhn S, Hegenbart U, Schönland SO, Wiese S, Read C, Schmidt M, Fändrich M. Role of mutations and post-translational modifications in systemic AL amyloidosis studied by cryo-EM. Nat Commun 2021; 12:6434. [PMID: 34741031 PMCID: PMC8571268 DOI: 10.1038/s41467-021-26553-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/01/2021] [Indexed: 12/29/2022] Open
Abstract
Systemic AL amyloidosis is a rare disease that is caused by the misfolding of immunoglobulin light chains (LCs). Potential drivers of amyloid formation in this disease are post-translational modifications (PTMs) and the mutational changes that are inserted into the LCs by somatic hypermutation. Here we present the cryo electron microscopy (cryo-EM) structure of an ex vivo λ1-AL amyloid fibril whose deposits disrupt the ordered cardiomyocyte structure in the heart. The fibril protein contains six mutational changes compared to the germ line and three PTMs (disulfide bond, N-glycosylation and pyroglutamylation). Our data imply that the disulfide bond, glycosylation and mutational changes contribute to determining the fibril protein fold and help to generate a fibril morphology that is able to withstand proteolytic degradation inside the body.
Collapse
Affiliation(s)
- Lynn Radamaker
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | | | - Giada Andreotti
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Julian Baur
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | | | - Sarah Schreiner
- Medical Department V, Section of Multiple Myeloma, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Natalie Berghaus
- Medical Department V, Section of Multiple Myeloma, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Raoul Motika
- Department of Asia-Africa-Studies, Middle Eastern History and Culture, University of Hamburg, 20148, Hamburg, Germany
| | - Christian Haupt
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Volker Schmidt
- Institute of Stochastics, Ulm University, 89081, Ulm, Germany
| | - Stefanie Huhn
- Medical Department V, Section of Multiple Myeloma, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Ute Hegenbart
- Medical Department V, Amyloidosis Center, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Stefan O Schönland
- Medical Department V, Amyloidosis Center, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Clarissa Read
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
- Institute of Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Matthias Schmidt
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
19
|
Xu L, Su Y. Genetic pathogenesis of immunoglobulin light chain amyloidosis: basic characteristics and clinical applications. Exp Hematol Oncol 2021; 10:43. [PMID: 34284823 PMCID: PMC8290569 DOI: 10.1186/s40164-021-00236-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/11/2021] [Indexed: 02/05/2023] Open
Abstract
Immunoglobulin light chain amyloidosis (AL) is an indolent plasma cell disorder characterized by free immunoglobulin light chain (FLC) misfolding and amyloid fibril deposition. The cytogenetic pattern of AL shows profound similarity with that of other plasma cell disorders but harbors distinct features. AL can be classified into two primary subtypes: non-hyperdiploidy and hyperdiploidy. Non-hyperdiploidy usually involves immunoglobulin heavy chain translocations, and t(11;14) is the hallmark of this disease. T(11;14) is associated with low plasma cell count but high FLC level and displays distinct response outcomes to different treatment modalities. Hyperdiploidy is associated with plasmacytosis and subclone formation, and it generally confers a neutral or inferior prognostic outcome. Other chromosome abnormalities and driver gene mutations are considered as secondary cytogenetic aberrations that occur during disease evolution. These genetic aberrations contribute to the proliferation of plasma cells, which secrete excess FLC for amyloid deposition. Other genetic factors, such as specific usage of immunoglobulin light chain germline genes and light chain somatic mutations, also play an essential role in amyloid fibril deposition in AL. This paper will propose a framework of AL classification based on genetic aberrations and discuss the amyloid formation of AL from a genetic aspect.
Collapse
Affiliation(s)
- Linchun Xu
- Shantou University Medical College, Shantou, 515031, Guangdong, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yongzhong Su
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
20
|
Schürmann J, Gottwald J, Rottenaicher G, Tholey A, Röcken C. MALDI mass spectrometry imaging unravels organ and amyloid-type specific peptide signatures in pulmonary and gastrointestinal amyloidosis. Proteomics Clin Appl 2021; 15:e2000079. [PMID: 34061454 DOI: 10.1002/prca.202000079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Amyloidosis is a disease group caused by pathological aggregation and deposition of peptides in diverse tissue sites. Recently, matrix-assisted laser desorption/ionization mass spectrometry imaging coupled with ion mobility separation (MALDI-IMS MSI) was introduced as a novel tool to identify and classify amyloidosis using single sections from formalin-fixed and paraffin-embedded cardiac biopsies. Here, we tested the hypothesis that MALDI-IMS MSI can be applied to lung and gastrointestinal specimens. EXPERIMENTAL DESIGN Forty six lung and 65 gastrointestinal biopsy and resection specimens with different types of amyloid were subjected to MALDI-IMS MSI. Ninety three specimens included tissue areas without amyloid as internal negative controls. Nine cases without amyloid served as additional negative controls. RESULTS Utilizing a peptide filter method and 21 known amyloid specific tryptic peptides we confirmed the applicability of a universal peptide signature with a sensitivity of 100% and a specificity of 100% for the detection of amyloid deposits in the lung and gastrointestinal tract. Additionally, the frequencies of individual m/z-values of the 21 tryptic marker peptides showed organ- and tissue-type specific differences. CONCLUSIONS AND CLINICAL RELEVANCE MALDI-IMS MSI adds a valuable analytical approach to diagnose and classify amyloid and the detection frequency of individual tryptic peptides is organ-/tissue-type specific.
Collapse
Affiliation(s)
- Jan Schürmann
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Juliane Gottwald
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Georg Rottenaicher
- Center for Integrated Protein Science Munich at the Department of Chemistry, Technical University of Munich, Garching, Germany
| | - Andreas Tholey
- Systematic Proteome Research & Bioanalytics, Institute of Experimental Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
21
|
Abstract
Light-chain amyloidosis (AL) is a fatal disorder wherein the immunoglobulin light chain misfolds and aggregates, leading to amyloid plaques in various organs. Patient-specific mutations in the light chain variable domain (VL) are tightly linked to amyloidosis, but how these mutations drive AL is unknown. In recent work, Rottenaicher et al. analyze five mutations found in the VL of a patient with cardiac AL. Their data suggest that decreased VL stability and increased flexibility in the core of the VL, caused by mutations outside of this core, could be key to aggregation and highlight the delicate balancing act required for antibody maturation to enable antigen recognition while not altering protein biophysics.
Collapse
|
22
|
Yan NL, Santos-Martins D, Nair R, Chu A, Wilson IA, Johnson KA, Forli S, Morgan GJ, Petrassi HM, Kelly JW. Discovery of Potent Coumarin-Based Kinetic Stabilizers of Amyloidogenic Immunoglobulin Light Chains Using Structure-Based Design. J Med Chem 2021; 64:6273-6299. [PMID: 33939422 DOI: 10.1021/acs.jmedchem.1c00339] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In immunoglobulin light-chain (LC) amyloidosis, transient unfolding or unfolding and proteolysis enable aggregation of LC proteins, causing potentially fatal organ damage. A drug that kinetically stabilizes LCs could suppress aggregation; however, LC sequences are variable and have no natural ligands, hindering drug development efforts. We previously identified high-throughput screening hits that bind to a site at the interface between the two variable domains of the LC homodimer. We hypothesized that extending the stabilizers beyond this initially characterized binding site would improve affinity. Here, using protease sensitivity assays, we identified stabilizers that can be divided into four substructures. Some stabilizers exhibit nanomolar EC50 values, a 3000-fold enhancement over the screening hits. Crystal structures reveal a key π-π stacking interaction with a conserved tyrosine residue that was not utilized by the screening hits. These data provide a foundation for developing LC stabilizers with improved binding selectivity and enhanced physicochemical properties.
Collapse
Affiliation(s)
- Nicholas L Yan
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Diogo Santos-Martins
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Reji Nair
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Alan Chu
- California Institute for Biomedical Research, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Kristen A Johnson
- California Institute for Biomedical Research, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Gareth J Morgan
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, United States.,The Amyloidosis Center, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - H Michael Petrassi
- California Institute for Biomedical Research, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States.,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|