1
|
Wu YW, Wei CH, Lin YH, Huang SY, Liu YW, Tan TD, Yu WC, Liu CJ, Hung CL, Ko PS, Yang NI, Cho SF, Huang CY, Liao CK, Jerry Teng CL, Chen TY, Chen WJ, Ko BS. Joint position statement of the diagnosis and management of light chain amyloidosis and light chain cardiac amyloidosis by the Taiwan Society of Cardiology (TSOC) and the Hematology Society of Taiwan (HST). J Formos Med Assoc 2025:S0929-6646(25)00217-7. [PMID: 40368759 DOI: 10.1016/j.jfma.2025.05.006] [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: 09/01/2024] [Revised: 03/31/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
Light chain (AL) amyloidosis, defined by the organ deposition of misfolded and aggregated amyloid light chain, is the most common subtype of systematic amyloidosis with an incidence rate of around 1 case per 100,000 person-years in Taiwan. Due to its rarity, and heterogeneous initial presentation depending on the affected organs, diagnosis and management of AL amyloidosis are challenging. To increase the awareness of AL amyloidosis across clinical specialties and provide evidence-based recommendations, the Taiwan Society of Cardiology (TSOC) and Hematology Society of Taiwan (HST) have jointly developed a position statement. The joint statement provides diagnosis and management recommendations for systematic AL amyloidosis and AL cardiac amyloidosis under Taiwan's medical context.
Collapse
Affiliation(s)
- Yen-Wen Wu
- Division of Cardiology, Cardiovascular Medical Center, Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
| | - Chao-Hung Wei
- Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yen-Hung Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shang-Yi Huang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yen-Wen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan; College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tran-Der Tan
- Department of Hematopoietic Stem Cell Transplantation and Cell Therapy, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Wen-Chung Yu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Jen Liu
- Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Lieh Hung
- Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, United States
| | - Po-Shen Ko
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Hematology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ning-I Yang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Feng Cho
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Yao Huang
- Division of Cardiology and Cardiovascular Research Center, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Chun-Kai Liao
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chieh-Lin Jerry Teng
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan; Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tsai-Yun Chen
- College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wen-Jone Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan.
| | - Bor-Sheng Ko
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan; Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
2
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
3
|
Arias-Alcala J, Novas-Moreno C, Novillo-Sarmiento C, Lopez-Lopez I, Soriano S, Martin-Malo A, Rodelo-Haad C. Beta-2 microglobulin-associated amyloidosis: A forgotten link to remember. Nefrologia 2025; 45:97-99. [PMID: 39757080 DOI: 10.1016/j.nefroe.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Affiliation(s)
| | | | | | - Isabel Lopez-Lopez
- Nephrology Service, University Hospital Reina Sofia, Cordoba-Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain University of Cordoba, Cordoba, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Sagrario Soriano
- Nephrology Service, University Hospital Reina Sofia, Cordoba-Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain University of Cordoba, Cordoba, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain; European Uremic Toxins Group (EUTOx)
| | - Alejandro Martin-Malo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain University of Cordoba, Cordoba, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain; European Uremic Toxins Group (EUTOx)
| | - Cristian Rodelo-Haad
- Nephrology Service, University Hospital Reina Sofia, Cordoba-Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain University of Cordoba, Cordoba, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain; European Uremic Toxins Group (EUTOx).
| |
Collapse
|
4
|
Ploper D, Pernicone AO, Tomas-Grau RH, Manzano VE, Socías SB, Teran MDM, Budeguer Isa V, Sosa-Padilla B, González-Lizárraga F, Avila CL, Guayán ML, Chaves S, Cruz H, Vera Pingitore E, Varela O, Chehín R. Design, Synthesis, and Evaluation of a Novel Conjugate Molecule with Dopaminergic and Neuroprotective Activities for Parkinson's Disease. ACS Chem Neurosci 2024; 15:2795-2810. [PMID: 38991155 DOI: 10.1021/acschemneuro.4c00169] [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] [Indexed: 07/13/2024] Open
Abstract
The escalating prevalence of Parkinson's disease (PD) underscores the need for innovative therapeutic interventions since current palliative measures, including the standard l-Dopa formulations, face challenges of tolerance and side effects while failing to address the underlying neurodegenerative processes. Here, we introduce DAD9, a novel conjugate molecule that aims to combine symptomatic relief with disease-modifying strategies for PD. Crafted through knowledge-guided chemistry, the molecule combines a nonantibiotic doxycycline derivative with dopamine, preserving neuroprotective attributes while maintaining dopaminergic agonism. This compound exhibited no off-target effects on PD-relevant cell functions and sustained antioxidant and anti-inflammatory properties of the tetracycline precursor. Furthermore, it effectively interfered with the formation and seeding of toxic α-synuclein aggregates without producing detrimental oxidative species. In addition, DAD9 was able to activate dopamine receptors, and docking simulations shed light onto the molecular details of this interaction. These findings position DAD9 as a potential neuroprotective dopaminergic agonist, promising advancements in PD therapeutics.
Collapse
Affiliation(s)
- Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Agustín O Pernicone
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Rodrigo H Tomas-Grau
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Verónica E Manzano
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Sergio B Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - María Del Milagro Teran
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Valentina Budeguer Isa
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Bernardo Sosa-Padilla
- Instituto de Química del Noroeste Argentino (INQUINOA) (CONICET), Universidad Nacional de Tucumán (UNT), Ayacucho 471, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - César L Avila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - María Laura Guayán
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Silvina Chaves
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Hernán Cruz
- Instituto de Química Física, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Lorenzo 456, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Esteban Vera Pingitore
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Oscar Varela
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| |
Collapse
|
5
|
Kaku T, Ikebukuro K, Tsukakoshi K. Structure of cytotoxic amyloid oligomers generated during disaggregation. J Biochem 2024; 175:575-585. [PMID: 38430131 PMCID: PMC11155694 DOI: 10.1093/jb/mvae023] [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: 08/30/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
Amyloidosis is characterized by the abnormal accumulation of amyloid proteins. The causative proteins aggregate from monomers to oligomers and fibrils, among which some intermediate oligomers are considered as major toxins. Cytotoxic oligomers are generated not only by aggregation but also via fibril disaggregation. However, little is known about the structural characteristics and generation conditions of cytotoxic oligomers produced during disaggregation. Herein, we summarized the structural commonalities of cytotoxic oligomers formed under various disaggregation conditions, including the addition of heat shock proteins or small compounds. In vitro experimental data demonstrated the presence of high-molecular-weight oligomers (protofibrils or protofilaments) that exhibited a fibrous morphology and β-sheet structure. Molecular dynamics simulations indicated that the distorted β-sheet structure contributed to their metastability. The tendency of these cytotoxic oligomers to appear under mild disaggregation conditions, implied formation during the early stages of disaggregation. This review will aid researchers in exploring the characteristics of highly cytotoxic oligomers and developing drugs that target amyloid aggregates.
Collapse
Affiliation(s)
- Toshisuke Kaku
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
6
|
Muthu SA, Qureshi A, Sharma R, Bisaria I, Parvez S, Grover S, Ahmad B. Redesigning the kinetics of lysozyme amyloid aggregation by cephalosporin molecules. J Biomol Struct Dyn 2024:1-16. [PMID: 38682862 DOI: 10.1080/07391102.2024.2335304] [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: 12/03/2023] [Accepted: 03/20/2024] [Indexed: 05/01/2024]
Abstract
In lysozyme amyloidosis, fibrillar aggregates of lysozyme are associated with severe renal, hepatic, and gastrointestinal manifestations, with no definite therapy. Current drugs are now being tested in amyloidosis clinical trials as aggregation inhibitors to mitigate disease progression. The tetracycline group among antimicrobials in use is in phase II of clinical trials, whereas some macrolides and cephalosporins have shown neuroprotection. In the present study, two cephalosporins, ceftazidime (CZD) and cefotaxime (CXM), and a glycopeptide, vancomycin (VNC), are evaluated for inhibition of amyloid aggregation of hen egg white lysozyme (HEWL) under two conditions (i) 4 M guanidine hydrochloride (GuHCl) at pH 6.5 and 37° C, (ii) At pH 1.5 and 65 °C. Fluorescence quench titration and molecular docking methods report that CZD, CXM, and VNC interact more strongly with the partially folded intermediates (PFI) in comparison to the protein's natural state (N). However, only CZD and CXM proficiently inhibit the aggregation. Transmission electron microscopy, tinctorial assessments, and aggregation kinetics all support oligomer-level inhibition. Transition structures in CZD-HEWL and CXM-HEWL aggregation are shown by circular dichroism (CD). On the other hand, kinetic variables and soluble fraction assays point to a localized association of monomers. Intrinsic fluorescence (IF),1-Anilino 8-naphthalene sulphonic acid, and CD demonstrate structural and conformational modifications redesigning the PFI. GuHCl-induced unfolding and differential scanning fluorimetry suggested that the PFI monomers bound to CZD and CXM exhibited partial stability. Our results present two mechanisms that function in both solution conditions, creating a novel avenue for the screening of putative inhibitors for drug repurposing. We extend our proposed mechanisms in the designing of physical inhibitors of amyloid aggregation considering shorter time frames and foolproof methods.
Collapse
Affiliation(s)
- Shivani A Muthu
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
- Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi, India
| | - Afnaan Qureshi
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Rahul Sharma
- Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi, India
| | - Ishita Bisaria
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Sonam Grover
- Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi, India
| | - Basir Ahmad
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
7
|
Tan M, Chen Y, Ooi M, de Mel S, Tan D, Soekojo C, Tso A, Khoo CY, Tan HZ, Choo J, Lee LK, Diong CP, Goh YT, Hwang W, Linn YC, Ho A, Chng WJ, Nagarajan C. AL amyloidosis: Singapore Myeloma Study Group consensus guidelines on diagnosis, treatment and management. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2023; 52:601-624. [PMID: 38920149 DOI: 10.47102/annals-acadmedsg.2023101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
AL amyloidosis is the most common form of systemic amyloidosis. However, the non-specific nature of presenting symptoms requires the need for a heightened clinical suspicion to detect unexplained manifestations in the appropriate clinical setting. Early detection and treatment are crucial as the degree of cardiac involvement emerges as a primary prognostic predictor of survival in a patient with AL amyloidosis. Following the diagnosis of AL amyloidosis with appropriate tissue biopsies, prompt treatment with a bortezomib, cyclophosphamide and dexamethasone-based first-line induction with or without daratumumab should be initiated. The goal of treatment is to achieve the best haematologic response possible, ideally with involved free light chain <20 mg/L, as it offers the best chance of organ function improvement. Treatment should be changed if patients do not achieve a partial response within 2 cycles of treatment or very good partial response after 4 cycles or after autologous stem cell transplant, as achievement of profound and prolonged clonal responses translates to better organ response and long-term outcomes. Early involvement of multidisciplinary subspecialists such as renal physicians, cardiologists, neurologists, and gastroenterologists for optimal maintenance and support of involved organs is recommended for optimal management of patients with AL amyloidosis.
Collapse
Affiliation(s)
- Melinda Tan
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Yunxin Chen
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Melissa Ooi
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Sanjay de Mel
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Daryl Tan
- Clinic for Lymphoma, Myeloma and Blood Disorders, Singapore
| | - Cinnie Soekojo
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Allison Tso
- Department of Haematology, Tan Tock Seng Hospital, Singapore
| | - Chun Yuan Khoo
- Department of Cardiology, National Heart Centre, Singapore
| | - Hui Zhuan Tan
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Jason Choo
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Lian King Lee
- Department of Haematology, Tan Tock Seng Hospital, Singapore
| | | | - Yeow Tee Goh
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - William Hwang
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Yeh Ching Linn
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Aloysius Ho
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Wee Joo Chng
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Chandramouli Nagarajan
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| |
Collapse
|
8
|
Sulatsky MI, Stepanenko OV, Stepanenko OV, Mikhailova EV, Kuznetsova IM, Turoverov KK, Sulatskaya AI. Amyloid fibrils degradation: the pathway to recovery or aggravation of the disease? Front Mol Biosci 2023; 10:1208059. [PMID: 37377863 PMCID: PMC10291066 DOI: 10.3389/fmolb.2023.1208059] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Background: The most obvious manifestation of amyloidoses is the accumulation of amyloid fibrils as plaques in tissues and organs, which always leads to a noticeable deterioration in the patients' condition and is the main marker of the disease. For this reason, early diagnosis of amyloidosis is difficult, and inhibition of fibrillogenesis, when mature amyloids are already accumulated in large quantities, is ineffective. A new direction for amyloidosis treatment is the development of approaches aimed at the degradation of mature amyloid fibrils. In the present work, we investigated possible consequences of amyloid's degradation. Methods: We analyzed the size and morphology of amyloid degradation products by transmission and confocal laser scanning microscopy, their secondary structure and spectral properties of aromatic amino acids, intrinsic chromophore sfGFP, and fibril-bound amyloid-specific probe thioflavin T (ThT) by the absorption, fluorescence and circular dichroism spectroscopy, as well as the cytotoxicity of the formed protein aggregates by MTT-test and their resistance to ionic detergents and boiling by SDS-PAGE. Results: On the example of sfGFP fibrils (model fibrils, structural rearrangements of which can be detected by a specific change in the spectral properties of their chromophore), and pathological Aβ-peptide (Aβ42) fibrils, leading to neuronal death in Alzheimer's disease, the possible mechanisms of amyloids degradation after exposure to factors of different nature (proteins with chaperone and protease activity, denaturant, and ultrasound) was demonstrated. Our study shows that, regardless of the method of fibril degradation, the resulting species retain some amyloid's properties, including cytotoxicity, which may even be higher than that of intact amyloids. Conclusion: The results of our work indicate that the degradation of amyloid fibrils in vivo should be treated with caution since such an approach can lead not to recovery, but to aggravation of the disease.
Collapse
Affiliation(s)
- Maksim I. Sulatsky
- Laboratory of cell morphology, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga V. Stepanenko
- Laboratory of structural dynamics, stability and folding of proteins, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Olesya V. Stepanenko
- Laboratory of structural dynamics, stability and folding of proteins, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Ekaterina V. Mikhailova
- Laboratory of structural dynamics, stability and folding of proteins, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Irina M. Kuznetsova
- Laboratory of structural dynamics, stability and folding of proteins, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Konstantin K. Turoverov
- Laboratory of structural dynamics, stability and folding of proteins, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Anna I. Sulatskaya
- Laboratory of structural dynamics, stability and folding of proteins, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
9
|
Acharya I, Dominah GA, Xing D, Allen E, Iding JS, Haas CJ. Peritoneal Amyloid as a Presenting Manifestation of AL Amyloid. J Community Hosp Intern Med Perspect 2023; 13:96-102. [PMID: 37877063 PMCID: PMC10593169 DOI: 10.55729/2000-9666.1193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 10/26/2023] Open
Abstract
Amyloid is a systemic disease characterized by extracellular deposition of misfolded protein. Gastrointestinal and peritoneal deposition of light chain (AL) amyloid is an under-recognized manifestation of this systemic disease, usually as a late sequela. Here we present a case of recently diagnosed AL peritoneal amyloid that presented in the context of recurrent, acute onset abdominal discomfort and was found to have bowel obstruction complicated by perforation in the setting of AL-mediated gastrointestinal tract infiltration and dysmotility.
Collapse
Affiliation(s)
- Indira Acharya
- Medstar Health Internal Medicine Residency Program, Baltimore, MD,
USA
| | - Gifty A. Dominah
- Medstar Health Internal Medicine Residency Program, Baltimore, MD,
USA
| | - Dongmei Xing
- Medstar Health, Department of Pathology, Baltimore, MD,
USA
| | | | | | | |
Collapse
|
10
|
Almeida ZL, Brito RMM. Amyloid Disassembly: What Can We Learn from Chaperones? Biomedicines 2022; 10:3276. [PMID: 36552032 PMCID: PMC9776232 DOI: 10.3390/biomedicines10123276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/23/2022] Open
Abstract
Protein aggregation and subsequent accumulation of insoluble amyloid fibrils with cross-β structure is an intrinsic characteristic of amyloid diseases, i.e., amyloidoses. Amyloid formation involves a series of on-pathway and off-pathway protein aggregation events, leading to mature insoluble fibrils that eventually accumulate in multiple tissues. In this cascade of events, soluble oligomeric species are formed, which are among the most cytotoxic molecular entities along the amyloid cascade. The direct or indirect action of these amyloid soluble oligomers and amyloid protofibrils and fibrils in several tissues and organs lead to cell death in some cases and organ disfunction in general. There are dozens of different proteins and peptides causing multiple amyloid pathologies, chief among them Alzheimer's, Parkinson's, Huntington's, and several other neurodegenerative diseases. Amyloid fibril disassembly is among the disease-modifying therapeutic strategies being pursued to overcome amyloid pathologies. The clearance of preformed amyloids and consequently the arresting of the progression of organ deterioration may increase patient survival and quality of life. In this review, we compiled from the literature many examples of chemical and biochemical agents able to disaggregate preformed amyloids, which have been classified as molecular chaperones, chemical chaperones, and pharmacological chaperones. We focused on their mode of action, chemical structure, interactions with the fibrillar structures, morphology and toxicity of the disaggregation products, and the potential use of disaggregation agents as a treatment option in amyloidosis.
Collapse
Affiliation(s)
| | - Rui M. M. Brito
- Chemistry Department and Coimbra Chemistry Centre—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
11
|
Prokaeva T, Joshi T, Klimtchuk ES, Gibson VM, Spencer B, Siddiqi O, Nedelkov D, Hu Y, Berk JL, Cuddy SAM, Dasari S, Chiu A, Choate LA, McPhail ED, Cui H, Chen H, Burks EJ, Sanchorawala V, Connors LH. A novel substitution of proline (P32L) destabilises β2-microglobulin inducing hereditary systemic amyloidosis. Amyloid 2022; 29:255-262. [PMID: 35575118 DOI: 10.1080/13506129.2022.2072199] [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: 01/05/2023]
Abstract
BACKGROUND β2-microglobulin amyloidosis was first described in the 1980s as a protein deposition disease associated with long-term haemodialysis. More recently, two inherited forms resulting from separate point mutations in the β2-microglobulin gene have been identified. In this report, we detail a novel β2M variant, P32L, caused by a unique dinucleotide mutation that is linked to systemic hereditary β2-microglobulin amyloidosis. METHODS Three family members from a Portuguese kinship featured cardiomyopathy, requiring organ transplantation in one case, along with soft tissue involvement; other involvements included gastrointestinal, neuropathic and sicca syndrome. In vitro studies with recombinant P32L, P32G, D76N and wild-type β2-microglobulin were undertaken to compare the biophysical properties of the proteins. RESULTS The P32L variant was caused by the unique heterozygous dinucleotide mutation c.154_155delinsTT. Amyloid disease featured lowered serum β2-microglobulin levels with near equal amounts of circulating P32L and wild-type proteins; amyloid deposits were composed exclusively of P32L variant protein. In vitro studies of P32L demonstrated thermodynamic and chemical instability and enhanced susceptibility to proteolysis with rapid formation of pre-fibrillar oligomeric structures by N- and C-terminally truncated species under physiological conditions. CONCLUSIONS This work provides both clinical and experimental evidence supporting the critical role of P32 residue replacement in β2M amyloid fibrillogenesis.
Collapse
Affiliation(s)
- Tatiana Prokaeva
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Tracy Joshi
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Elena S Klimtchuk
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Victoria M Gibson
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Brian Spencer
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Omar Siddiqi
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | | | | | - John L Berk
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Sarah A M Cuddy
- Amyloidosis Program, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - April Chiu
- Department of Laboratory of Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lauren A Choate
- Department of Laboratory of Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ellen D McPhail
- Department of Laboratory of Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Haili Cui
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Eric J Burks
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Lawreen H Connors
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
12
|
Maya-Martinez R, Xu Y, Guthertz N, Walko M, Karamanos TK, Sobott F, Breeze AL, Radford SE. Dimers of D76N-β 2-microglobulin display potent antiamyloid aggregation activity. J Biol Chem 2022; 298:102659. [PMID: 36328246 PMCID: PMC9712992 DOI: 10.1016/j.jbc.2022.102659] [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: 06/09/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 11/05/2022] Open
Abstract
Self-association of WT β2-microglobulin (WT-β2m) into amyloid fibrils is associated with the disorder dialysis related amyloidosis. In the familial variant D76N-β2m, the single amino acid substitution enhances the aggregation propensity of the protein dramatically and gives rise to a disorder that is independent of renal dysfunction. Numerous biophysical and structural studies on WT- and D76N-β2m have been performed in order to better understand the structure and dynamics of the native proteins and their different potentials to aggregate into amyloid. However, the structural properties of transient D76N-β2m oligomers and their role(s) in assembly remained uncharted. Here, we have utilized NMR methods, combined with photo-induced crosslinking, to detect, trap, and structurally characterize transient dimers of D76N-β2m. We show that the crosslinked D76N-β2m dimers have different structures from those previously characterized for the on-pathway dimers of ΔN6-β2m and are unable to assemble into amyloid. Instead, the crosslinked D76N-β2m dimers are potent inhibitors of amyloid formation, preventing primary nucleation and elongation/secondary nucleation when added in substoichiometric amounts with D76N-β2m monomers. The results highlight the specificity of early protein-protein interactions in amyloid formation and show how mapping these interfaces can inform new strategies to inhibit amyloid assembly.
Collapse
Affiliation(s)
- Roberto Maya-Martinez
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Yong Xu
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nicolas Guthertz
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Martin Walko
- Astbury Centre for Structural Molecular Biology, School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - Theodoros K Karamanos
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Frank Sobott
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Alexander L Breeze
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
13
|
Velander P, Wu L, Hildreth SB, Vogelaar NJ, Mukhopadhyay B, Helm RF, Zhang S, Xu B. Catechol-containing compounds are a broad class of protein aggregation inhibitors: Redox state is a key determinant of the inhibitory activities. Pharmacol Res 2022; 184:106409. [PMID: 35995346 PMCID: PMC10074477 DOI: 10.1016/j.phrs.2022.106409] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/20/2022] [Accepted: 08/17/2022] [Indexed: 12/30/2022]
Abstract
A range of neurodegenerative and related aging diseases, such as Alzheimer's disease and type 2 diabetes, are linked to toxic protein aggregation. Yet the mechanisms of protein aggregation inhibition by small molecule inhibitors remain poorly understood, in part because most protein targets of aggregation assembly are partially unfolded or intrinsically disordered, which hinders detailed structural characterization of protein-inhibitor complexes and structural-based inhibitor design. Herein we employed a parallel small molecule library-screening approach to identify inhibitors against three prototype amyloidogenic proteins in neurodegeneration and related proteinopathies: amylin, Aβ and tau. One remarkable class of inhibitors identified from these screens against different amyloidogenic proteins was catechol-containing compounds and redox-related quinones/anthraquinones. Secondary assays validated most of the identified inhibitors. In vivo efficacy evaluation of a selected catechol-containing compound, rosmarinic acid, demonstrated its strong mitigating effects of amylin amyloid deposition and related diabetic pathology in transgenic HIP rats. Further systematic investigation of selected class of inhibitors under aerobic and anaerobic conditions revealed that the redox state of the broad class of catechol-containing compounds is a key determinant of the amyloid inhibitor activities. The molecular insights we gained not only explain why a large number of catechol-containing polyphenolic natural compounds, often enriched in healthy diet, have anti-neurodegeneration and anti-aging activities, but also could guide the rational design of therapeutic or nutraceutical strategies to target a broad range of neurodegenerative and related aging diseases.
Collapse
Affiliation(s)
- Paul Velander
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Ling Wu
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; BRITE Research Institute and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA; Affiliated Faculty, Duke/UNC Alzheimer's Disease Research Center, Durham, NC 27710, USA
| | - Sherry B Hildreth
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Nancy J Vogelaar
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; Center for Drug Discovery, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Biswarup Mukhopadhyay
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Richard F Helm
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Bin Xu
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; Center for Drug Discovery, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; BRITE Research Institute and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA; Affiliated Faculty, Duke/UNC Alzheimer's Disease Research Center, Durham, NC 27710, USA.
| |
Collapse
|
14
|
Tomas-Grau R, González-Lizárraga F, Ploper D, Avila CL, Socías SB, Besnault P, Tourville A, Mella RM, Villacé P, Salado C, Rose C, Seon-Méniel B, Brunel JM, Ferrié L, Raisman-Vozari R, Michel PP, Figadère B, Chehín R. Neuroprotective Effects of a Novel Demeclocycline Derivative Lacking Antibiotic Activity: From a Hit to a Promising Lead Compound. Cells 2022; 11:cells11172759. [PMID: 36078167 PMCID: PMC9454755 DOI: 10.3390/cells11172759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/09/2022] Open
Abstract
The antibiotic tetracycline demeclocycline (DMC) was recently reported to rescue α-synuclein (α-Syn) fibril-induced pathology. However, the antimicrobial activity of DMC precludes its potential use in long-term neuroprotective treatments. Here, we synthesized a doubly reduced DMC (DDMC) derivative with residual antibiotic activity and improved neuroprotective effects. The molecule was obtained by removal the dimethylamino substituent at position 4 and the reduction of the hydroxyl group at position 12a on ring A of DMC. The modifications strongly diminished its antibiotic activity against Gram-positive and Gram-negative bacteria. Moreover, this compound preserved the low toxicity of DMC in dopaminergic cell lines while improving its ability to interfere with α-Syn amyloid-like aggregation, showing the highest effectiveness of all tetracyclines tested. Likewise, DDMC demonstrated the ability to reduce seeding induced by the exogenous addition of α-Syn preformed fibrils (α-SynPFF) in biophysical assays and in a SH-SY5Y-α-Syn-tRFP cell model. In addition, DDMC rendered α-SynPFF less inflammogenic. Our results suggest that DDMC may be a promising drug candidate for hit-to-lead development and preclinical studies in Parkinson's disease and other synucleinopathies.
Collapse
Affiliation(s)
- Rodrigo Tomas-Grau
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - César L. Avila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Sergio B. Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Pierre Besnault
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Aurore Tourville
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Rosa M. Mella
- Innoprot SL, Parque Tecnológico de Bizkaia, Edificio 502, 48160 Derio, Spain
| | - Patricia Villacé
- Innoprot SL, Parque Tecnológico de Bizkaia, Edificio 502, 48160 Derio, Spain
| | - Clarisa Salado
- Innoprot SL, Parque Tecnológico de Bizkaia, Edificio 502, 48160 Derio, Spain
| | - Clémence Rose
- BioCIS, Université Paris-Saclay, CNRS, 92290 Châtenay-Malabry, France
| | | | - Jean-Michel Brunel
- UMR_MD1 “Membranes et Cibles Thérapeutiques”, U1261 INSERM, Aix-Marseille Université, 13385 Marseille, France
| | - Laurent Ferrié
- BioCIS, Université Paris-Saclay, CNRS, 92290 Châtenay-Malabry, France
| | - Rita Raisman-Vozari
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Patrick P. Michel
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Bruno Figadère
- BioCIS, Université Paris-Saclay, CNRS, 92290 Châtenay-Malabry, France
- Correspondence: (B.F.); (R.C.)
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
- Correspondence: (B.F.); (R.C.)
| |
Collapse
|
15
|
The potential use of tetracyclines in neurodegenerative diseases and the role of nano-based drug delivery systems. Eur J Pharm Sci 2022; 175:106237. [PMID: 35710076 DOI: 10.1016/j.ejps.2022.106237] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/07/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022]
Abstract
Neurodegenerative diseases are still a challenge for effective treatments. The high cost of approved drugs, severity of side effects, injection site pain, and restrictions on drug delivery to the Central Nervous System (CNS) can overshadow the management of these diseases. Due to the chronic and progressive evolution of neurodegenerative disorders and since there is still no cure for them, new therapeutic strategies such as the combination of several drugs or the use of existing drugs with new therapeutic applications are valuable strategies. Tetracyclines are traditionally classified as antibiotics. However, in this class of drugs, doxycycline and minocycline exhibit also anti-inflammatory effects by inhibiting microglia/macrophages. Hence, they have been studied as potential agents for the treatment of neurodegenerative diseases. The results of in vitro and in vivo studies confirm the effective role of these two drugs as anti-inflammatory agents in experimentally induced models of neurodegenerative diseases. In clinical studies, satisfactory results have been obtained in Multiple sclerosis (MS) but not yet in other disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), or Amyotrophic lateral sclerosis (ALS). In recent years, researchers have developed and evaluated nanoparticulate drug delivery systems to improve the clinical efficacy of these two tetracyclines for their potential application in neurodegenerative diseases. This study reviews the neuroprotective roles of minocycline and doxycycline in four of the main neurodegenerative disorders: AD, PD, ALS and MS. Moreover, the potential applications of nanoparticulate delivery systems developed for both tetracyclines are also reviewed.
Collapse
|
16
|
Markulin I, Matasin M, Turk VE, Salković-Petrisic M. Challenges of repurposing tetracyclines for the treatment of Alzheimer's and Parkinson's disease. J Neural Transm (Vienna) 2022; 129:773-804. [PMID: 34982206 DOI: 10.1007/s00702-021-02457-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
The novel antibiotic-exploiting strategy in the treatment of Alzheimer's (AD) and Parkinson's (PD) disease has emerged as a potential breakthrough in the field. The research in animal AD/PD models provided evidence on the antiamyloidogenic, anti-inflammatory, antioxidant and antiapoptotic activity of tetracyclines, associated with cognitive improvement. The neuroprotective effects of minocycline and doxycycline in animals initiated investigation of their clinical efficacy in AD and PD patients which led to inconclusive results and additionally to insufficient safety data on a long-standing doxycycline and minocycline therapy in these patient populations. The safety issues should be considered in two levels; in AD/PD patients (particularly antibiotic-induced alteration of gut microbiota and its consequences), and as a world-wide threat of development of bacterial resistance to these antibiotics posed by a fact that AD and PD are widespread incurable diseases which require daily administered long-lasting antibiotic therapy. Recently proposed subantimicrobial doxycycline doses should be thoroughly explored for their effectiveness and long-term safety especially in AD/PD populations. Keeping in mind the antibacterial activity-related far-reaching undesirable effects both for the patients and globally, further work on repurposing these drugs for a long-standing therapy of AD/PD should consider the chemically modified tetracycline compounds tailored to lack antimicrobial but retain (or introduce) other activities effective against the AD/PD pathology. This strategy might reduce the risk of long-term therapy-related adverse effects (particularly gut-related ones) and development of bacterial resistance toward the tetracycline antibiotic agents but the therapeutic potential and desirable safety profile of such compounds in AD/PD patients need to be confirmed.
Collapse
Affiliation(s)
- Iva Markulin
- Community Health Centre Zagreb-Centre, Zagreb, Croatia
| | | | - Viktorija Erdeljic Turk
- Division of Clinical Pharmacology, Department of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Melita Salković-Petrisic
- Department of Pharmacology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia.
| |
Collapse
|
17
|
Mitochondria and Antibiotics: For Good or for Evil? Biomolecules 2021; 11:biom11071050. [PMID: 34356674 PMCID: PMC8301944 DOI: 10.3390/biom11071050] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 01/16/2023] Open
Abstract
The discovery and application of antibiotics in the common clinical practice has undeniably been one of the major medical advances in our times. Their use meant a drastic drop in infectious diseases-related mortality and contributed to prolonging human life expectancy worldwide. Nevertheless, antibiotics are considered by many a double-edged sword. Their extensive use in the past few years has given rise to a global problem: antibiotic resistance. This factor and the increasing evidence that a wide range of antibiotics can damage mammalian mitochondria, have driven a significant sector of the medical and scientific communities to advise against the use of antibiotics for purposes other to treating severe infections. Notwithstanding, a notorious number of recent studies support the use of these drugs to treat very diverse conditions, ranging from cancer to neurodegenerative or mitochondrial diseases. In this context, there is great controversy on whether the risks associated to antibiotics outweigh their promising beneficial features. The aim of this review is to provide insight in the topic, purpose for which the most relevant findings regarding antibiotic therapies have been discussed.
Collapse
|
18
|
Morgan GJ. Barriers to Small Molecule Drug Discovery for Systemic Amyloidosis. Molecules 2021; 26:3571. [PMID: 34208058 PMCID: PMC8230685 DOI: 10.3390/molecules26123571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
Inhibition of amyloid fibril formation could benefit patients with systemic amyloidosis. In this group of diseases, deposition of amyloid fibrils derived from normally soluble proteins leads to progressive tissue damage and organ failure. Amyloid formation is a complex process, where several individual steps could be targeted. Several small molecules have been proposed as inhibitors of amyloid formation. However, the exact mechanism of action for a molecule is often not known, which impedes medicinal chemistry efforts to develop more potent molecules. Furthermore, commonly used assays are prone to artifacts that must be controlled for. Here, potential mechanisms by which small molecules could inhibit aggregation of immunoglobulin light-chain dimers, the precursor proteins for amyloid light-chain (AL) amyloidosis, are studied in assays that recapitulate different aspects of amyloidogenesis in vitro. One molecule reduced unfolding-coupled proteolysis of light chains, but no molecules inhibited aggregation of light chains or disrupted pre-formed amyloid fibrils. This work demonstrates the challenges associated with drug development for amyloidosis, but also highlights the potential to combine therapies that target different aspects of amyloidosis.
Collapse
Affiliation(s)
- Gareth J Morgan
- Section of Hematology and Medical Oncology, Amyloidosis Center, Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
19
|
Obici L, Adams D. Acquired and inherited amyloidosis: Knowledge driving patients' care. J Peripher Nerv Syst 2021; 25:85-101. [PMID: 32378274 DOI: 10.1111/jns.12381] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/19/2022]
Abstract
Until recently, systemic amyloidoses were regarded as ineluctably disabling and life-threatening diseases. However, this field has witnessed major advances in the last decade, with significant improvements in therapeutic options and in the availability of accurate and non-invasive diagnostic tools. Outstanding progress includes unprecedented hematological response rates provided by risk-adapted regimens in light chain (AL) amyloidosis and the approval of innovative pharmacological agents for both hereditary and wild-type transthyretin amyloidosis (ATTR). Moreover, the incidence of secondary (AA) amyloidosis has continuously reduced, reflecting advances in therapeutics and overall management of several chronic inflammatory diseases. The identification and validation of novel therapeutic targets has grounded on a better knowledge of key molecular events underlying protein misfolding and aggregation and on the increasing availability of diagnostic, prognostic and predictive markers of organ damage and response to treatment. In this review, we focus on these recent advancements and discuss how they are translating into improved outcomes. Neurological involvement dominates the clinical picture in transthyretin and gelsolin inherited amyloidosis and has a significant impact on disease course and management in all patients. Neurologists, therefore, play a major role in improving patients' journey to diagnosis and in providing early access to treatment in order to prevent significant disability and extend survival.
Collapse
Affiliation(s)
- Laura Obici
- Amyloidosis Research and Treatment Centre, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - David Adams
- National Reference Center for Familial Amyloid Polyneuropathy and Other Rare Neuropathies, APHP, Université Paris Saclay, INSERM U1195, Le Kremlin Bicêtre, France
| |
Collapse
|
20
|
Muchtar E, Dispenzieri A, Gertz MA, Kumar SK, Buadi FK, Leung N, Lacy MQ, Dingli D, Ailawadhi S, Bergsagel PL, Fonseca R, Hayman SR, Kapoor P, Grogan M, Abou Ezzeddine OF, Rosenthal JL, Mauermann M, Siddiqui M, Gonsalves WI, Kourelis TV, Larsen JT, Reeder CB, Warsame R, Go RS, Murray DL, McPhail ED, Dasari S, Jevremovic D, Kyle RA, Lin Y, Lust JA, Russell SJ, Hwa YL, Fonder AL, Hobbs MA, Rajkumar SV, Roy V, Sher T. Treatment of AL Amyloidosis: Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART) Consensus Statement 2020 Update. Mayo Clin Proc 2021; 96:1546-1577. [PMID: 34088417 DOI: 10.1016/j.mayocp.2021.03.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/31/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022]
Abstract
Immunoglobulin light chain (AL) amyloidosis is a clonal plasma cell disorder leading to progressive and life-threatening organ failure. The heart and the kidneys are the most commonly involved organs, but almost any organ can be involved. Because of the nonspecific presentation, diagnosis delay is common, and many patients are diagnosed with advanced organ failure. In the era of effective therapies and improved outcomes for patients with AL amyloidosis, the importance of early recognition is further enhanced as the ability to reverse organ dysfunction is limited in those with a profound organ failure. As AL amyloidosis is an uncommon disorder and given patients' frailty and high early death rate, management of this complex condition is challenging. The treatment of AL amyloidosis is based on various anti-plasma cell therapies. These therapies are borrowed and customized from the treatment of multiple myeloma, a more common disorder. However, a growing number of phase 2/3 studies dedicated to the AL amyloidosis population are being performed, making treatment decisions more evidence-based. Supportive care is an integral part of management of AL amyloidosis because of the inherent organ dysfunction, limiting the delivery of effective therapy. This extensive review brings an updated summary on the management of AL amyloidosis, sectioned into the 3 pillars for survival improvement: early disease recognition, anti-plasma cell therapy, and supportive care.
Collapse
Affiliation(s)
- Eli Muchtar
- Division of Hematology, Mayo Clinic, Rochester, MN.
| | | | | | | | | | - Nelson Leung
- Division of Hematology, Mayo Clinic, Rochester, MN; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | | | - David Dingli
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | - Rafael Fonseca
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ
| | | | | | - Martha Grogan
- Division of Hematology, Mayo Clinic, Rochester, MN; Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | - Jeremy T Larsen
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ
| | - Craig B Reeder
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ
| | | | - Ronald S Go
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ellen D McPhail
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Dragan Jevremovic
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - John A Lust
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Yi Lisa Hwa
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | - S Vincent Rajkumar
- Division of Hematology, Mayo Clinic, Rochester, MN; Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Vivek Roy
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | - Taimur Sher
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
21
|
Mori W, Yuzu K, Lobsiger N, Nishioka H, Sato H, Nagase T, Iwaya K, Lindgren M, Zako T. Degradation of insulin amyloid by antibiotic minocycline and formation of toxic intermediates. Sci Rep 2021; 11:6857. [PMID: 33767265 PMCID: PMC7994847 DOI: 10.1038/s41598-021-86001-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/08/2021] [Indexed: 12/28/2022] Open
Abstract
Insulin balls, localized insulin amyloids formed at subcutaneous insulin-injection sites in patients with diabetes, cause poor glycemic control owing to impairments in insulin absorption. Our previous study has shown that some insulin balls are cytotoxic, but others are not, implying amyloid polymorphism. Interestingly, the patient with toxic insulin balls had been treated with antibiotic minocycline, suggesting a possible relationship between toxicity of insulin balls and minocycline. However, the direct effect of minocycline on the structure and cytotoxicity of the insulin amyloid is still unclear. Herein, we demonstrated that that minocycline at physiological concentrations induced degradation of insulin amyloids formed from human insulin and insulin drug preparations used for diabetes patients. Interestingly, the process involved the initial appearance of the toxic species, which subsequently changed into less-toxic species. It is also shown that the structure of the toxic species was similar to that of sonicated fragments of human insulin amyloids. Our study shed new light on the clarification of the revelation of insulin balls and the development of the insulin analogs for diabetes therapy.
Collapse
Affiliation(s)
- Wakako Mori
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, 790-8577, Japan
| | - Keisuke Yuzu
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, 790-8577, Japan
| | - Nadine Lobsiger
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, 790-8577, Japan
- Institute for Chemical and Bioengineering, ETH Zürich, 8093, Zürich, Switzerland
| | - Hideo Nishioka
- Application Management Department, JEOL Ltd, Tokyo, 196-8558, Japan
| | - Hisako Sato
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, 790-8577, Japan
| | - Terumasa Nagase
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, 3000395, Japan
| | - Keiichi Iwaya
- Department of Pathology, SASAKI Institute, Kyoundo Hospital, Tokyo, 101-0062, Japan
| | - Mikael Lindgren
- Department of Physics, Faculty of Natural Sciences, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Tamotsu Zako
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, 790-8577, Japan.
| |
Collapse
|
22
|
Vijayan D, Chandra R. Amyloid Beta Hypothesis in Alzheimer's Disease: Major Culprits and Recent Therapeutic Strategies. Curr Drug Targets 2021; 21:148-166. [PMID: 31385768 DOI: 10.2174/1389450120666190806153206] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/13/2019] [Accepted: 07/26/2019] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is one of the most common forms of dementia and has been a global concern for several years. Due to the multi-factorial nature of the disease, AD has become irreversible, fatal and imposes a tremendous socio-economic burden. Even though experimental medicines suggested moderate benefits, AD still lacks an effective treatment strategy for the management of symptoms or cure. Among the various hypotheses that describe development and progression of AD, the amyloid hypothesis has been a long-term adherent to the AD due to the involvement of various forms of Amyloid beta (Aβ) peptides in the impairment of neuronal and cognitive functions. Hence, majority of the drug discovery approaches in the past have focused on the prevention of the accumulation of Aβ peptides. Currently, there are several agents in the phase III clinical trials that target Aβ or the various macromolecules triggering Aβ deposition. In this review, we present the state of the art knowledge on the functional aspects of the key players involved in the amyloid hypothesis. Furthermore, we also discuss anti-amyloid agents present in the Phase III clinical trials.
Collapse
Affiliation(s)
- Dileep Vijayan
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Remya Chandra
- Department of Biotechnology and Microbiology, Thalassery Campus, Kannur University, Kerala Pin 670 661, India
| |
Collapse
|
23
|
Dominguez-Meijide A, Parrales V, Vasili E, González-Lizárraga F, König A, Lázaro DF, Lannuzel A, Haik S, Del Bel E, Chehín R, Raisman-Vozari R, Michel PP, Bizat N, Outeiro TF. Doxycycline inhibits α-synuclein-associated pathologies in vitro and in vivo. Neurobiol Dis 2021; 151:105256. [PMID: 33429042 DOI: 10.1016/j.nbd.2021.105256] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are neurodegenerative disorders characterized by the misfolding and aggregation of alpha-synuclein (aSyn). Doxycycline, a tetracyclic antibiotic shows neuroprotective effects, initially proposed to be due to its anti-inflammatory properties. More recently, an additional mechanism by which doxycycline may exert its neuroprotective effects has been proposed as it has been shown that it inhibits amyloid aggregation. Here, we studied the effects of doxycycline on aSyn aggregation in vivo, in vitro and in a cell free system using real-time quaking induced conversion (RT-QuiC). Using H4, SH-SY5Y and HEK293 cells, we found that doxycycline decreases the number and size of aSyn aggregates in cells. In addition, doxycycline inhibits the aggregation and seeding of recombinant aSyn, and attenuates the production of mitochondrial-derived reactive oxygen species. Finally, we found that doxycycline induces a cellular redistribution of aggregates in a C.elegans animal model of PD, an effect that is associated with a recovery of dopaminergic function. In summary, we provide strong evidence that doxycycline treatment may be an effective strategy against synucleinopathies.
Collapse
Affiliation(s)
- Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany; Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Valeria Parrales
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Eftychia Vasili
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | | | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Diana F Lázaro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Annie Lannuzel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France; University Hospital of Pointe-à-Pitre, Neurology Department, route de Chauvel, 97139 Abymes, Guadeloupe
| | - Stéphane Haik
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France; AP-HP, Cellule Nationale de Référence des Maladies de Creutzfeldt-Jakob, University Hospital Pitié-Salpêtrière, Paris F-75013, France
| | - Elaine Del Bel
- Department of Basic and Oral Biology, Faculty of Odontology of Ribeirão Preto, University of São Paulo (USP), Av do Café s/n, São Paulo, Brazil
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Argentina
| | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Patrick P Michel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Nicolas Bizat
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France; Faculté de Pharmacie de Paris, Paris University, 4 avenue de l'Observatoire, Paris F-75006, France.
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany; Max Planck Institute for Experimental Medicine, Goettingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK.
| |
Collapse
|
24
|
González-Lizárraga F, Ploper D, Ávila CL, Socías SB, Dos-Santos-Pereira M, Machín B, Del-Bel E, Michel PP, Pietrasanta LI, Raisman-Vozari R, Chehín R. CMT-3 targets different α-synuclein aggregates mitigating their toxic and inflammogenic effects. Sci Rep 2020; 10:20258. [PMID: 33219264 PMCID: PMC7679368 DOI: 10.1038/s41598-020-76927-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder for which only symptomatic treatments are available. Repurposing drugs that target α-synuclein aggregation, considered one of the main drivers of PD progression, could accelerate the development of disease-modifying therapies. In this work, we focused on chemically modified tetracycline 3 (CMT-3), a derivative with reduced antibiotic activity that crosses the blood–brain barrier and is pharmacologically safe. We found that CMT-3 inhibited α-synuclein amyloid aggregation and led to the formation of non-toxic molecular species, unlike minocycline. Furthermore, CMT-3 disassembled preformed α-synuclein amyloid fibrils into smaller fragments that were unable to seed in subsequent aggregation reactions. Most interestingly, disaggregated species were non-toxic and less inflammogenic on brain microglial cells. Finally, we modelled the interactions between CMT-3 and α-synuclein aggregates by molecular simulations. In this way, we propose a mechanism for fibril disassembly. Our results place CMT-3 as a potential disease modifier for PD and possibly other synucleinopathies.
Collapse
Affiliation(s)
- Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - César L Ávila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Sergio B Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | | | - Belén Machín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Elaine Del-Bel
- Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Patrick Pierre Michel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Lía I Pietrasanta
- Departamento de Física-Instituto de Física de Buenos Aires (IFIBA, UBA-CONICET) and Centro de Microscopías Avanzadas (CMA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France.
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina.
| |
Collapse
|
25
|
Insights into a Protein-Nanoparticle System by Paramagnetic Perturbation NMR Spectroscopy. MOLECULES (BASEL, SWITZERLAND) 2020; 25:molecules25215187. [PMID: 33171781 PMCID: PMC7664681 DOI: 10.3390/molecules25215187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/12/2020] [Accepted: 10/22/2020] [Indexed: 11/18/2022]
Abstract
Background: The interaction between proteins and nanoparticles is a very relevant subject because of the potential applications in medicine and material science in general. Further interest derives from the amyloidogenic character of the considered protein, β2-microglobulin (β2m), which may be regarded as a paradigmatic system for possible therapeutic strategies. Previous evidence showed in fact that gold nanoparticles (AuNPs) are able to inhibit β2m fibril formation in vitro. Methods: NMR (Nuclear Magnetic Resonance) and ESR (Electron Spin Resonance) spectroscopy are employed to characterize the paramagnetic perturbation of the extrinsic nitroxide probe Tempol on β2m in the absence and presence of AuNPs to determine the surface accessibility properties and the occurrence of chemical or conformational exchange, based on measurements conducted under magnetization equilibrium and non-equilibrium conditions. Results: The nitroxide perturbation analysis successfully identifies the protein regions where protein-protein or protein-AuNPs interactions hinder accessibility or/and establish exchange contacts. These information give interesting clues to recognize the fibrillation interface of β2m and hypothesize a mechanism for AuNPs fibrillogenesis inhibition. Conclusions: The presented approach can be advantageously applied to the characterization of the interface in protein-protein and protein-nanoparticles interactions.
Collapse
|
26
|
Wiglenda T, Groenke N, Hoffmann W, Manz C, Diez L, Buntru A, Brusendorf L, Neuendorf N, Schnoegl S, Haenig C, Schmieder P, Pagel K, Wanker EE. Sclerotiorin Stabilizes the Assembly of Nonfibrillar Abeta42 Oligomers with Low Toxicity, Seeding Activity, and Beta-sheet Content. J Mol Biol 2020; 432:2080-2098. [PMID: 32061932 DOI: 10.1016/j.jmb.2020.01.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/14/2020] [Accepted: 01/28/2020] [Indexed: 01/21/2023]
Abstract
The self-assembly of the 42-residue amyloid-β peptide, Aβ42, into fibrillar aggregates is associated with neuronal dysfunction and toxicity in Alzheimer's disease (AD) patient brains, suggesting that small molecules acting on this process might interfere with pathogenesis. Here, we present experimental evidence that the small molecule sclerotiorin (SCL), a natural product belonging to the group of azaphilones, potently delays both seeded and nonseeded Aβ42 polymerization in cell-free assays. Mechanistic biochemical studies revealed that the inhibitory effect of SCL on fibrillogenesis is caused by its ability to kinetically stabilize small Aβ42 oligomers. These structures exhibit low β-sheet content and do not possess seeding activity, indicating that SCL acts very early in the amyloid formation cascade before the assembly of seeding-competent, β-sheet-rich fibrillar aggregates. Investigations with NMR WaterLOGSY experiments confirmed the association of Aβ42 assemblies with SCL in solution. Furthermore, using ion mobility-mass spectrometry, we observed that SCL directly interacts with a small fraction of Aβ42 monomers in the gas phase. In comparison to typical amyloid fibrils, small SCL-stabilized Aβ42 assemblies are inefficiently taken up into mammalian cells and have low toxicity in cell-based assays. Overall, these mechanistic studies support a pathological role of stable, β-sheet-rich Aβ42 fibrils in AD, while structures with low β-sheet content may be less relevant.
Collapse
Affiliation(s)
- Thomas Wiglenda
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Nicole Groenke
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Waldemar Hoffmann
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Christian Manz
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Lisa Diez
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Alexander Buntru
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Lydia Brusendorf
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Nancy Neuendorf
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sigrid Schnoegl
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christian Haenig
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Peter Schmieder
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Kevin Pagel
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Erich E Wanker
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
27
|
Faravelli G, Raimondi S, Marchese L, Partridge FA, Soria C, Mangione PP, Canetti D, Perni M, Aprile FA, Zorzoli I, Di Schiavi E, Lomas DA, Bellotti V, Sattelle DB, Giorgetti S. C. elegans expressing D76N β 2-microglobulin: a model for in vivo screening of drug candidates targeting amyloidosis. Sci Rep 2019; 9:19960. [PMID: 31882874 PMCID: PMC6934621 DOI: 10.1038/s41598-019-56498-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/10/2019] [Indexed: 01/16/2023] Open
Abstract
The availability of a genetic model organism with which to study key molecular events underlying amyloidogenesis is crucial for elucidating the mechanism of the disease and the exploration of new therapeutic avenues. The natural human variant of β2-microglobulin (D76N β2-m) is associated with a fatal familial form of systemic amyloidosis. Hitherto, no animal model has been available for studying in vivo the pathogenicity of this protein. We have established a transgenic C. elegans line, expressing the human D76N β2-m variant. Using the INVertebrate Automated Phenotyping Platform (INVAPP) and the algorithm Paragon, we were able to detect growth and motility impairment in D76N β2-m expressing worms. We also demonstrated the specificity of the β2-m variant in determining the pathological phenotype by rescuing the wild type phenotype when β2-m expression was inhibited by RNA interference (RNAi). Using this model, we have confirmed the efficacy of doxycycline, an inhibitor of the aggregation of amyloidogenic proteins, in rescuing the phenotype. In future, this C. elegans model, in conjunction with the INVAPP/Paragon system, offers the prospect of high-throughput chemical screening in the search for new drug candidates.
Collapse
Affiliation(s)
- Giulia Faravelli
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy.
| | - Sara Raimondi
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy
| | - Loredana Marchese
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy
| | - Frederick A Partridge
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, Gower Street, London, WC1E 6JF, United Kingdom
| | - Cristina Soria
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy
| | - P Patrizia Mangione
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, University College London, London, UK
| | - Diana Canetti
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, University College London, London, UK
| | - Michele Perni
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Francesco A Aprile
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Irene Zorzoli
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and Bioresources (IBBR), CNR, 80131, Naples, Italy
| | - David A Lomas
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, Gower Street, London, WC1E 6JF, United Kingdom
| | - Vittorio Bellotti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, University College London, London, UK
| | - David B Sattelle
- Centre for Respiratory Biology, UCL Respiratory, Division of Medicine, University College London, Gower Street, London, WC1E 6JF, United Kingdom
| | - Sofia Giorgetti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100, Pavia, Italy.
| |
Collapse
|
28
|
Marcinko TM, Liang C, Savinov S, Chen J, Vachet RW. Structural Heterogeneity in the Preamyloid Oligomers of β-2-Microglobulin. J Mol Biol 2019; 432:396-409. [PMID: 31711963 DOI: 10.1016/j.jmb.2019.10.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 11/29/2022]
Abstract
In dialysis patients, the protein β2-microglobulin (β2m) forms amyloid fibrils in a condition known as dialysis-related amyloidosis. To understand the early stages of the amyloid assembly process, we have used native electrospray ionization (ESI) together with ion mobility mass spectrometry (IM-MS) to study soluble preamyloid oligomers. ESI-IM-MS reveals the presence of multiple conformers for the dimer, tetramer, and hexamer that precede the Cu(II)-induced amyloid assembly process, results which are distinct from β2m oligomers formed at low pH. Experimental and computational results indicate that the predominant dimer is a Cu(II)-bound structure with an antiparallel side-by-side configuration. In contrast, tetramers exist in solution in both Cu(II)-bound and Cu(II)-free forms. Selective depletion of Cu(II)-bound species results in two primary conformers-one that is compact and another that is more expanded. Molecular modeling and molecular dynamics simulations identify models for these two tetrameric conformers with unique interactions and interfaces that enthalpically compensate for the loss of Cu(II). Unlike with other amyloid systems in which conformational heterogeneity is often associated with different amyloid morphologies or off-pathway events, conformational heterogeneity in the tetramer seems to be a necessary aspect of Cu(II)-induced amyloid formation by β2m. Moreover, the Cu(II)-free models represent a new advance in our understanding of Cu(II) release in Cu(II)-induced amyloid formation, laying a foundation for further mechanistic studies as well as development of new inhibition strategies.
Collapse
Affiliation(s)
- Tyler M Marcinko
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States
| | - Chungwen Liang
- Computational and Modeling Core Facility, Institute for Applied Life Sciences, Amherst, MA 01003, United States
| | - Sergey Savinov
- Computational and Modeling Core Facility, Institute for Applied Life Sciences, Amherst, MA 01003, United States; Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, United States
| | - Jianhen Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States; Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, United States
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
29
|
The Anti-Amyloidogenic Action of Doxycycline: A Molecular Dynamics Study on the Interaction with Aβ42. Int J Mol Sci 2019; 20:ijms20184641. [PMID: 31546787 PMCID: PMC6769662 DOI: 10.3390/ijms20184641] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/11/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
The pathological aggregation of amyloidogenic proteins is a hallmark of many neurological diseases, including Alzheimer’s disease and prion diseases. We have shown both in vitro and in vivo that doxycycline can inhibit the aggregation of Aβ42 amyloid fibrils and disassemble mature amyloid fibrils. However, the molecular mechanisms of the drug’s anti-amyloidogenic property are not understood. In this study, a series of molecular dynamics simulations were performed to explain the molecular mechanism of the destabilization of Aβ42 fibrils by doxycycline and to compare the action of doxycycline with those of iododoxorubicin (a toxic structural homolog of tetracyclines), curcumin (known to have anti-amyloidogenic activity) and gentamicin (an antibiotic with no experimental evidence of anti-amyloidogenic properties). We found that doxycycline tightly binds the exposed hydrophobic amino acids of the Aβ42 amyloid fibrils, partly leading to destabilization of the fibrillar structure. Clarifying the molecular determinants of doxycycline binding to Aβ42 may help devise further strategies for structure-based drug design for Alzheimer’s disease.
Collapse
|
30
|
Santagati G, Cataldo E, Columbano V, Chatrenet A, Penna D, Pelosi E, Hachemi M, Gendrot L, Nielsen L, Cinquantini F, Saulnier P, Arena V, Boursot C, Piccoli GB. Positron Emission Tomography Can Support the Diagnosis of Dialysis-Related Amyloidosis. J Clin Med 2019; 8:jcm8091494. [PMID: 31546847 PMCID: PMC6781261 DOI: 10.3390/jcm8091494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/31/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The improvements in dialysis have not eliminated long-term problems, including dialysis-related amyloidosis (DRA), caused by Beta-2 microglobulin deposition. Several types of scintigraphy have been tested to detect DRA, none entered the clinical practice. Aim of the study was to assess the potential of PET-FDG scan in the diagnosis of DRA. METHODS Forty-six dialysis patients with at least one PET scan (72 scans) were selected out 162 patients treated in 2016-2018. Subjective global assessment (SGA), malnutrition inflammation score (A), Charlson Comorbidity Index (CCI), were assessed at time of scan; 218 age-matched cases with normal kidney function were selected as controls. PET scans were read in duplicate. Carpal tunnel syndrome was considered a proxy for DRA. A composite "amyloid score" score considered each dialysis year = 1 point; carpal tunnel-DRA = 5 points per site. Logistic regression, ROC curves and a prediction model were built. RESULTS The prevalence of positive PET was 43.5% in dialysis, 5% in controls (p < 0.0001). PET was positive in 14/15 (93.3%) scans in patients with carpal tunnel. PET sensitivity for detecting DRA was 95% (specificity 64%). Carpal tunnel was related to dialysis vintage and MIS. A positive PET scan was significantly associated with dialysis vintage, MIS and amyloid score. A prediction model to explain PET positivity combined clinical score and MIS, allowing for an AUC of 0.906 (CI: 0.813-0.962; p < 0.001). CONCLUSIONS PET-FDG may identify DRA, and may be useful in detecting cases in which inflammation favours B2M deposition. This finding, needing large-scale confirmation, could open new perspectives in the study of DRA.
Collapse
Affiliation(s)
| | | | | | | | - Daniele Penna
- Affidea IRMET, PET CENTER, Torino via Onorato Vigliani 89, 10135 Torino, Italy.
| | - Ettore Pelosi
- Affidea IRMET, PET CENTER, Torino via Onorato Vigliani 89, 10135 Torino, Italy.
| | - Mammar Hachemi
- Medecine Nucleaire, Centre Hospitalier du Mans, 72037 Le Mans, France.
| | | | - Louise Nielsen
- Néphrologie, Centre Hospitalier du Mans, 72037 Le Mans, France.
| | | | | | - Vincenzo Arena
- Affidea IRMET, PET CENTER, Torino via Onorato Vigliani 89, 10135 Torino, Italy.
| | - Charles Boursot
- Medecine Nucleaire, Centre Hospitalier du Mans, 72037 Le Mans, France.
| | - Giorgina Barbara Piccoli
- Néphrologie, Centre Hospitalier du Mans, 72037 Le Mans, France.
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, 10100 Torino, Italy.
| |
Collapse
|
31
|
Iwaya K, Zako T, Fukunaga J, Sörgjerd KM, Ogata K, Kogure K, Kosano H, Noritake M, Maeda M, Ando Y, Katsura Y, Nagase T. Toxicity of insulin-derived amyloidosis: a case report. BMC Endocr Disord 2019; 19:61. [PMID: 31196059 PMCID: PMC6567432 DOI: 10.1186/s12902-019-0385-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Insulin-derived amyloidosis is a skin-related complication of insulin therapy that interferes with insulin therapy. Although toxicities of in vitro-formed insulin amyloid fibrils have been well studied, the toxicity of insulin-derived amyloidosis remains to be clarified. CASE PRESENTATION A 58-year-old man with type 2 diabetes mellitus underwent a lower limb amputation due to diabetic gangrene. Several antibiotics including minocycline were administered for infection and sepsis. A hard mass at the insulin injection sites in the lower abdomen was discovered by chance four months later. Although no abnormal findings in the surface skin of the mass were observed, necrotic tissue was seen around the mass when a biopsy was performed. Histological and toxicity studies were performed for this patient and four other patients with abdominal masses at insulin injection sites. Histological and immunohistochemical studies showed that the masses had typical characteristics of amyloid deposits in all cases, whereas necrotic findings were seen adjacent to the amyloid deposit only in the case presented. Toxicity studies indicated that the amyloid tissue from the present case had significant cell toxicity compared to the control skin tissue or the amyloid tissues from the other four cases. CONCLUSIONS This report showed that toxic insulin-derived amyloidosis can occur. In addition, this report suggested that toxic insulin-derived amyloidosis may cause necrosis in the surrounding tissue. Although the toxic amyloid deposit of insulin-derived amyloidosis was found in only one patient, no structural differences between toxic and non-toxic deposits were seen on histological and immunohistochemical studies.
Collapse
Affiliation(s)
- Keiichi Iwaya
- Department of Pathology, SASAKI Institute, Kyoundo Hospital, Tokyo, Japan
| | - Tamotsu Zako
- Bioengineering Laboratory, RIKEN Cluster for Pioneering Research, Saitama, Japan
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, Japan
| | - Junta Fukunaga
- Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Ehime, Japan
| | | | - Kentaro Ogata
- Department of Pathology, Tachikawa Hospital, Tokyo, Japan
| | - Koichiro Kogure
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Ibaraki, 300-0395 Japan
| | - Hiroshi Kosano
- Faculty of Pharmaceutical Science, Teikyo University, Tokyo, Japan
| | - Masayuki Noritake
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Ibaraki, 300-0395 Japan
| | - Mizuo Maeda
- Bioengineering Laboratory, RIKEN Cluster for Pioneering Research, Saitama, Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshiya Katsura
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Ibaraki, 300-0395 Japan
| | - Terumasa Nagase
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Ibaraki, 300-0395 Japan
| |
Collapse
|
32
|
Puscalau-Girtu I, Scheller JS, Claus S, Fändrich M. Cell assay for the identification of amyloid inhibitors in systemic AA amyloidosis. Amyloid 2019; 26:24-33. [PMID: 30739503 DOI: 10.1080/13506129.2019.1568978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Systemic AA amyloidosis is still, up to this day, a life-threatening complication of chronic inflammatory diseases. Despite the success of anti-inflammatory treatment, the prognosis of some AA patients is still poor, which is why therapies directed at the amyloidogenic pathway in AA amyloidosis are being sought after. The cell culture model of amyloid formation from serum amyloid A1 (SAA1) protein remodels crucial features of AA amyloid deposit formation in vivo. We here demonstrate how the cell model can be utilized for the identification of compounds with amyloid inhibitory activity. Out of five compounds previously reported to inhibit self-assembly of various amyloidogenic proteins, we found that epigallocatechin gallate (EGCG) inhibited the formation of SAA1-derived fibrils in cell culture. From a series of compounds targeting the protein quality control machinery, the autophagy inhibitor wortmannin reduced amyloid formation, while the other tested compounds did not lead to a substantial reduction of the amyloid load. These data suggest that amyloid formation can be targeted not only via the protein self-assembly pathway directly, but also by treatment with compounds that impact the cellular protein machinery.
Collapse
Affiliation(s)
| | | | - Stephanie Claus
- a Institute of Protein Biochemistry, Ulm University , Ulm , Germany
| | - Marcus Fändrich
- a Institute of Protein Biochemistry, Ulm University , Ulm , Germany
| |
Collapse
|
33
|
Gamir-Morralla A, Sacristán S, Medina M, Iglesias T. Effects of Thioflavin T and GSK-3 Inhibition on Lifespan and Motility in a Caenorhabditis elegans Model of Tauopathy. J Alzheimers Dis Rep 2019; 3:47-57. [PMID: 30842997 PMCID: PMC6400111 DOI: 10.3233/adr-180087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The nematode Caenorhabditis elegans (C. elegans) is a powerful model organism to study lifespan and aging, protein aggregation, and neurodegeneration, as well as to carry out drug screenings. The C. elegans strain aex-3/T337 expresses human pathogenic V337M mutant tau under a pan-neuronal promoter and presents uncoordinated locomotion, accumulation of phosphorylated insoluble tau, and shortened lifespan. Herein we have used this strain to assay two compounds that could affect tau aggregation and/or phosphorylation, and looked for phenotypic changes in their lifespan and motility. The first compound is Thioflavin T (ThT), a member of the tetracycline family with protein antiaggregant properties, yet to be tested in a tauopathy model. The second is a novel small molecule, NP103, a highly selective inhibitor of glycogen synthase kinase-3 (GSK-3), the main kinase contributing to pathogenic tau hyperphosphorylation. Importantly, we find that ThT extends lifespan of aex-3/T337 worms as it does with control N2 animals, showing both strains similar locomotion features under this treatment. By contrast, NP103 improves the paralysis phenotype of aex-3/T337 mutants but not their lifespan. Our results show that both treatments present beneficial effects for this model of tauopathy and encourage pursuing further investigations on their therapeutic potential for AD and other tauopathies.
Collapse
Affiliation(s)
- Andrea Gamir-Morralla
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Sacristán
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Miguel Medina
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
34
|
Amigoni L, Airoldi C, Natalello A, Romeo M, Diomede L, Tortora P, Regonesi ME. Methacycline displays a strong efficacy in reducing toxicity in a SCA3 Caenorhabditis elegans model. Biochim Biophys Acta Gen Subj 2019; 1863:279-290. [DOI: 10.1016/j.bbagen.2018.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 11/28/2022]
|
35
|
CMV-Specific Immune Response-New Patients, New Insight: Central Role of Specific IgG during Infancy and Long-Lasting Immune Deficiency after Allogenic Stem Cell Transplantation. Int J Mol Sci 2019; 20:ijms20020271. [PMID: 30641912 PMCID: PMC6358762 DOI: 10.3390/ijms20020271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/21/2022] Open
Abstract
Although the existing paradigm states that cytomegalovirus (CMV) reactivation is under the control of the cellular immune response, the role of humoral and innate counterparts are underestimated. The study analyzed the host–virus interaction i.e., CMV-immune response evolution during infection in three different clinical situations: (1) immunodeficient CMV-positive human leukocyte antigen (HLA)-matched bone marrow recipients after immunoablative conditioning as well as immunocompetent, (2) adult, and (3) infant with primary immune response. In the first situation, a fast and significant decrease of specific immunity was observed but reconstitution of marrow-derived B and natural killer (NK) cells was observed prior to thymic origin of T cells. The lowest CMV-IgG (93.2 RU/mL) was found just before CMV viremia. It is noteworthy that the sole and exclusive factor of CMV-specific immune response is a residual recipient antibody class IgG. The CMV-quantiferon increase was detected later, but in the first phase, phytohemagglutinin (PHA)-induced IFN-γ release was significantly lower than that of CMV-induced (“indeterminate” results). It corresponds with the increase of NK cells at the top of lymphocyte reconstitution and undetected CMV-specific CD8 cells using a pentamer technique. In immunocompetent adult (CMV-negative donor), the cellular and humoral immune response increased in a parallel manner, but symptoms of CMV mononucleosis persisted until the increase of specific IgG. During infancy, the decrease of the maternal CMV-IgG level to 89.08 RU/mL followed by clinical sequel, i.e., CMV replication, were described. My observations shed light on a unique host-CMV interaction and CMV-IgG role: they indicate that its significant decrease predicts CMV replication. Before primary cellular immune response development, the high level of residual CMV-IgG (about >100 R/mL) from mother or recipient prevents virus reactivation. The innate immune response and NK-dependent IFN-secretion should be further investigated.
Collapse
|
36
|
Lucchetti J, Fracasso C, Balducci C, Passoni A, Forloni G, Salmona M, Gobbi M. Plasma and Brain Concentrations of Doxycycline after Single and Repeated Doses in Wild-Type and APP23 Mice. J Pharmacol Exp Ther 2019; 368:32-40. [PMID: 30396916 DOI: 10.1124/jpet.118.252064] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/26/2018] [Indexed: 03/08/2025] Open
Abstract
Repurposing doxycycline for the treatment of amyloidosis has recently been put forward because of the antiaggregating and anti-inflammatory properties of the drug. Most of the investigations of the therapeutic potential of doxycycline for neurodegenerative amyloidosis, e.g., prion and Alzheimer disease (AD), have been carried out in mouse models, but surprisingly no data are available regarding the concentrations reached in the brain after systemic administration. We filled this gap by analyzing the pharmacokinetic profile of doxycycline in plasma and brain after single and repeated intraperitoneal injections of 10 and 100 mg/kg, in wild-type mice and the APP23 mouse model of AD. The main outcomes of our study are: 1) Peak plasma concentrations ranged from 2 to10 μg/ml, superimposable to those in humans; 2) brain-to-plasma ratio was ∼0.2, comparable to the cerebrospinal fluid/serum ratios in humans; 3) brain Cmax 4-6 hours after a single dose was ∼0.5 (10 mg/kg) and ∼5 μM (100 mg/kg). Notably, these concentrations are lower than those required for the drug's antiaggregating properties as observed in cell-free studies, suggesting that other features underlie the positive cognitive effects in AD mice; 4) elimination half-life was shorter than in humans (3-6 vs. 15-30 hours), therefore no significant accumulation was observed in mouse brain following repeated treatments; and 5) there were no differences between doxycycline concentrations in brain areas of age-matched wild-type and APP23 mice. These data are useful for planning preclinical studies with translational validity, and to identify more reliably the mechanism(s) of action underlying the central in vivo effects of doxycycline.
Collapse
Affiliation(s)
- Jacopo Lucchetti
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Claudia Fracasso
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Claudia Balducci
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alice Passoni
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gianluigi Forloni
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mario Salmona
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Departments of Molecular Biochemistry and Pharmacology (J.L., C.F., M.S., M.G.), Neuroscience (C.B., G.F.), and Environmental Health Science (A.P.), Instituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
37
|
Giorgetti S, Greco C, Tortora P, Aprile FA. Targeting Amyloid Aggregation: An Overview of Strategies and Mechanisms. Int J Mol Sci 2018; 19:E2677. [PMID: 30205618 PMCID: PMC6164555 DOI: 10.3390/ijms19092677] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/02/2018] [Accepted: 09/05/2018] [Indexed: 12/26/2022] Open
Abstract
Amyloids result from the aggregation of a set of diverse proteins, due to either specific mutations or promoting intra- or extra-cellular conditions. Structurally, they are rich in intermolecular β-sheets and are the causative agents of several diseases, both neurodegenerative and systemic. It is believed that the most toxic species are small aggregates, referred to as oligomers, rather than the final fibrillar assemblies. Their mechanisms of toxicity are mostly mediated by aberrant interactions with the cell membranes, with resulting derangement of membrane-related functions. Much effort is being exerted in the search for natural antiamyloid agents, and/or in the development of synthetic molecules. Actually, it is well documented that the prevention of amyloid aggregation results in several cytoprotective effects. Here, we portray the state of the art in the field. Several natural compounds are effective antiamyloid agents, notably tetracyclines and polyphenols. They are generally non-specific, as documented by their partially overlapping mechanisms and the capability to interfere with the aggregation of several unrelated proteins. Among rationally designed molecules, we mention the prominent examples of β-breakers peptides, whole antibodies and fragments thereof, and the special case of drugs with contrasting transthyretin aggregation. In this framework, we stress the pivotal role of the computational approaches. When combined with biophysical methods, in several cases they have helped clarify in detail the protein/drug modes of interaction, which makes it plausible that more effective drugs will be developed in the future.
Collapse
Affiliation(s)
- Sofia Giorgetti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy.
| | - Claudio Greco
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126 Milano, Italy.
| | - Paolo Tortora
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy.
- Milan Center for Neuroscience (Neuro-MI), 20126 Milano, Italy.
| | - Francesco Antonio Aprile
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.
| |
Collapse
|
38
|
Brancolini G, Maschio MC, Cantarutti C, Corazza A, Fogolari F, Bellotti V, Corni S, Esposito G. Citrate stabilized gold nanoparticles interfere with amyloid fibril formation: D76N and ΔN6 β2-microglobulin variants. NANOSCALE 2018; 10:4793-4806. [PMID: 29469914 DOI: 10.1039/c7nr06808e] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Protein aggregation including the formation of dimers and multimers in solution, underlies an array of human diseases such as systemic amyloidosis which is a fatal disease caused by misfolding of native globular proteins damaging the structure and function of affected organs. Different kind of interactors can interfere with the formation of protein dimers and multimers in solution. A very special class of interactors are nanoparticles thanks to the extremely efficient extension of their interaction surface. In particular citrate-coated gold nanoparticles (cit-AuNPs) were recently investigated with amyloidogenic protein β2-microglobulin (β2m). Here we present the computational studies on two challenging models known for their enhanced amyloidogenic propensity, namely ΔN6 and D76N β2m naturally occurring variants, and disclose the role of cit-AuNPs on their fibrillogenesis. The proposed interaction mechanism lies in the interference of the cit-AuNPs with the protein dimers at the early stages of aggregation, that induces dimer disassembling. As a consequence, natural fibril formation can be inhibited. Relying on the comparison between atomistic simulations at multiple levels (enhanced sampling molecular dynamics and Brownian dynamics) and protein structural characterisation by NMR, we demonstrate that the cit-AuNPs interactors are able to inhibit protein dimer assembling. As a consequence, the natural fibril formation is also inhibited, as found in experiment.
Collapse
Affiliation(s)
- Giorgia Brancolini
- Center S3, CNR Institute Nanoscience, Via Campi 213/A, 41125 Modena, Italy.
| | | | - Cristina Cantarutti
- Dipartimento di Scienza Mediche e Biologiche (DSMB), University of Udine, Piazzale Kolbe 3, 33100 Udine, Italy
| | - Alessandra Corazza
- Dipartimento di Scienza Mediche e Biologiche (DSMB), University of Udine, Piazzale Kolbe 3, 33100 Udine, Italy and Istituto Nazionale Biostrutture e Biosistemi, Viale medaglie d'Oro, 305 - 00136 Roma, Italy
| | - Federico Fogolari
- Dipartimento di Scienza Mediche e Biologiche (DSMB), University of Udine, Piazzale Kolbe 3, 33100 Udine, Italy and Istituto Nazionale Biostrutture e Biosistemi, Viale medaglie d'Oro, 305 - 00136 Roma, Italy
| | - Vittorio Bellotti
- Dipartimento di Medicina Molecolare, Universita' di Pavia, Via Taramelli 3, 27100 Pavia, Italy and Istituto Nazionale Biostrutture e Biosistemi, Viale medaglie d'Oro, 305 - 00136 Roma, Italy and Division of Medicine, University College of London, London NW3 2PF, UK
| | - Stefano Corni
- Department of Chemical Science, University of Padova, via VIII Febbraio 2, 35122 Padova and Center S3, CNR Institute Nanoscience, Via Campi 213/A, 41125 Modena, Italy
| | - Gennaro Esposito
- Center S3, CNR Institute Nanoscience, Via Campi 213/A, 41125 Modena, Italy. and Istituto Nazionale Biostrutture e Biosistemi, Viale medaglie d'Oro, 305 - 00136 Roma, Italy and Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
39
|
Cantarutti C, Raj G, Fogolari F, Giorgetti S, Corazza A, Bellotti V, Naumov P, Esposito G. Interference of citrate-stabilized gold nanoparticles with β2-microglobulin oligomeric association. Chem Commun (Camb) 2018; 54:5422-5425. [DOI: 10.1039/c8cc01053f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Citrate-coated gold nanoparticles interfere with the association equilibria of β2-microglobulin and thus inhibit the early events of fibrillogenesis.
Collapse
Affiliation(s)
| | - Gijo Raj
- New York University Abu Dhabi
- Abu Dhabi
- United Arab Emirates
| | | | - Sofia Giorgetti
- Dipartimento di Medicina Molecolare
- Università di Pavia
- 27100 Pavia
- Italy
| | | | - Vittorio Bellotti
- Dipartimento di Medicina Molecolare
- Università di Pavia
- 27100 Pavia
- Italy
- Division of Medicine
| | - Panče Naumov
- New York University Abu Dhabi
- Abu Dhabi
- United Arab Emirates
| | - Gennaro Esposito
- New York University Abu Dhabi
- Abu Dhabi
- United Arab Emirates
- INBB
- 00136 Roma
| |
Collapse
|
40
|
Socias SB, González-Lizárraga F, Avila CL, Vera C, Acuña L, Sepulveda-Diaz JE, Del-Bel E, Raisman-Vozari R, Chehin RN. Exploiting the therapeutic potential of ready-to-use drugs: Repurposing antibiotics against amyloid aggregation in neurodegenerative diseases. Prog Neurobiol 2017; 162:17-36. [PMID: 29241812 DOI: 10.1016/j.pneurobio.2017.12.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 01/02/2023]
Abstract
Neurodegenerative diseases are chronic and progressive disorders that affect specific regions of the brain, causing gradual disability and suffering that results in a complete inability of patients to perform daily functions. Amyloid aggregation of specific proteins is the most common biological event that is responsible for neuronal death and neurodegeneration in various neurodegenerative diseases. Therapeutic agents capable of interfering with the abnormal aggregation are required, but traditional drug discovery has fallen short. The exploration of new uses for approved drugs provides a useful alternative to fill the gap between the increasing incidence of neurodegenerative diseases and the long-term assessment of classical drug discovery technologies. Drug re-profiling is currently the quickest possible transition from bench to bedside. In this way, experimental evidence shows that some antibiotic compounds exert neuroprotective action through anti-aggregating activity on disease-associated proteins. The finding that many antibiotics can cross the blood-brain barrier and have been used for several decades without serious toxic effects makes them excellent candidates for therapeutic switching towards neurological disorders. The present review is, to our knowledge, the first extensive evaluation and analysis of the anti-amyloidogenic effect of different antibiotics on well-known disease-associated proteins. In addition, we propose a common structural signature derived from the antiaggregant antibiotic molecules that could be relevant to rational drug discovery.
Collapse
Affiliation(s)
- Sergio B Socias
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI, San Miguel de Tucumán, Argentina, Argentina
| | - Florencia González-Lizárraga
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI, San Miguel de Tucumán, Argentina, Argentina
| | - Cesar L Avila
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI, San Miguel de Tucumán, Argentina, Argentina
| | - Cecilia Vera
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI, San Miguel de Tucumán, Argentina, Argentina
| | - Leonardo Acuña
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI, San Miguel de Tucumán, Argentina, Argentina; Sorbonne Universite, UPMC Univ Paris 06, INSERM, CNRS, UM75, U1127, UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| | - Julia E Sepulveda-Diaz
- Sorbonne Universite, UPMC Univ Paris 06, INSERM, CNRS, UM75, U1127, UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| | - Elaine Del-Bel
- Department of Morphology, Physiology and Stomatology, Faculty of Odontology of Ribeirão Preto, University of São Paulo, Brazil; Center of Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, Brazil
| | - Rita Raisman-Vozari
- Sorbonne Universite, UPMC Univ Paris 06, INSERM, CNRS, UM75, U1127, UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France.
| | - Rosana N Chehin
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI, San Miguel de Tucumán, Argentina, Argentina.
| |
Collapse
|
41
|
Liu T, Marcinko TM, Kiefer PA, Vachet RW. Using Covalent Labeling and Mass Spectrometry To Study Protein Binding Sites of Amyloid Inhibiting Molecules. Anal Chem 2017; 89:11583-11591. [PMID: 29028328 PMCID: PMC5677544 DOI: 10.1021/acs.analchem.7b02915] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Amyloid aggregates are associated with several debilitating diseases, and there are numerous efforts to develop small molecule treatments against these diseases. One challenge associated with these efforts is determining protein binding site information for potential therapeutics because amyloid-forming proteins rapidly form oligomers and aggregates, making traditional protein structural analysis techniques challenging. Using β-2-microglobulin (β2m) as a model amyloid-forming protein along with two recently identified small molecule amyloid inhibitors (i.e., rifamycin SV and doxycycline), we demonstrate that covalent labeling and mass spectrometry (MS) can be used to map small-molecule binding sites for a rapidly aggregating protein. Specifically, three different covalent labeling reagents, namely diethylpyrocarbonate, 2,3-butanedione, and the reagent pair EDC/GEE, are used together to pinpoint the binding sites of rifamycin SV, doxycycline, and another molecule, suramin, which binds but does not inhibit Cu(II)-induced β2m amyloid formation. The labeling results reveal binding sites that are consistent with the known effects of these molecules on β2m amyloid formation and are in general agreement with molecular docking results. We expect that this combined covalent labeling approach will be applicable to other protein/small molecule systems that are difficult to study by traditional means.
Collapse
Affiliation(s)
- Tianying Liu
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Tyler M. Marcinko
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Patrick A. Kiefer
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
42
|
Lessons learned from protein aggregation: toward technological and biomedical applications. Biophys Rev 2017; 9:501-515. [PMID: 28905328 DOI: 10.1007/s12551-017-0317-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 08/08/2017] [Indexed: 12/21/2022] Open
Abstract
The close relationship between protein aggregation and neurodegenerative diseases has been the driving force behind the renewed interest in a field where biophysics, neurobiology and nanotechnology converge in the study of the aggregate state. On one hand, knowledge of the molecular principles that govern the processes of protein aggregation has a direct impact on the design of new drugs for high-incidence pathologies that currently can only be treated palliatively. On the other hand, exploiting the benefits of protein aggregation in the design of new nanomaterials could have a strong impact on biotechnology. Here we review the contributions of our research group on novel neuroprotective strategies developed using a purely biophysical approach. First, we examine how doxycycline, a well-known and innocuous antibiotic, can reshape α-synuclein oligomers into non-toxic high-molecular-weight species with decreased ability to destabilize biological membranes, affect cell viability and form additional toxic species. This mechanism can be exploited to diminish the toxicity of α-synuclein oligomers in Parkinson's disease. Second, we discuss a novel function in proteostasis for extracellular glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in combination with a specific glycosaminoglycan (GAG) present in the extracellular matrix. GAPDH, by changing its quaternary structure from a tetramer to protofibrillar assembly, can kidnap toxic species of α-synuclein, and thereby interfere with the spreading of the disease. Finally, we review a brighter side of protein aggregation, that of exploiting the physicochemical advantages of amyloid aggregates as nanomaterials. For this, we designed a new generation of insoluble biocatalysts based on the binding of photo-immobilized enzymes onto hybrid protein:GAG amyloid nanofibrils. These new nanomaterials can be easily functionalized by attaching different enzymes through dityrosine covalent bonds.
Collapse
|
43
|
Doxycycline treatment in dialysis related amyloidosis: discrepancy between antalgic effect and inflammation, studied with FDG-positron emission tomography: a case report. BMC Nephrol 2017; 18:285. [PMID: 28874122 PMCID: PMC5586015 DOI: 10.1186/s12882-017-0698-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/23/2017] [Indexed: 12/24/2022] Open
Abstract
Background No effective treatment is currently available and dialysis related amyloidosis continues to be invalidating in long-term dialysis patients. A recent case series reported reduction of osteoarticular pain on doxycycline treatment, extending the indications of this drug, used in other uncommon forms of amyloidosis, to dialysis patients. Explanations of the antalgic effect were the anti-inflammatory properties and anti-coiling effects of tetracycline. Case presentation Our report regards a 54-year-old woman, who was never transplanted and has been on hemodialysis and hemodiafiltration for overall 37 years, due to renal hypoplasia. In spite of high efficiency hemodiafiltration, she complained of increasing, invalidating osteoarticular pain; history and imaging suggested beta-2 microglobulin amyloid. Positron emission tomography (PET scan) identified metabolically active lesions in the involved settings. Low-dose doxycycline (100 mg/day) was started, leading to a considerable decrease in pain (over 6 months, from 7 to 8 to 4–5 on a 0–10 scale). At 6 months, a PET scan showed unmodified or increased uptake in the involved settings. Conclusions In summary, the previously described antalgic effect of doxycycline in dialysis related amyloidosis is confirmed in our case, the first studied using PET scan. The pattern at PET can suggests that the antalgic effect is independent from inflammation and points to other factors, such as interaction with fibril geometry or with bone structure.
Collapse
|
44
|
Rationale and design of DUAL study: Doxycycline to Upgrade response in light chain (AL) amyloidosis (DUAL): A phase 2 pilot study of a two-pronged approach of prolonged doxycycline with plasma cell-directed therapy in the treatment of AL amyloidosis. Contemp Clin Trials Commun 2017; 8:33-38. [PMID: 29696194 PMCID: PMC5898504 DOI: 10.1016/j.conctc.2017.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 12/19/2022] Open
Abstract
Light chain (AL) amyloidosis is a plasma cell neoplasm associated with insoluble fibril deposition from clonal immunoglobulin chains systemically. The disease is associated with high early mortality and morbidity owing to advanced organ deposition as well as lack of proven de-fibrillogenic therapies. Pre-clinical and retrospective clinical data suggests that doxycycline has benefit in AL amyloidosis. The ongoing DUAL study is a single center, open label, phase 2 study in which patients with AL amyloidosis who are undergoing clone-directed therapy for the underlying neoplasm with oral doxycycline given for 1 year to test the hypothesis that prolonged doxycycline use will be safe, feasible, and lead to reduced early mortality in systemic AL amyloidosis and hasten organ amyloid response. Clinical follow up visits will occur at monthly intervals for systemic AL patients and at 3 monthly intervals for localized AL patients. Blood tests will be collected during these time points for hematologic response assessment. Organ testing will be conducted at 3 monthly intervals and radiologic testing will be conducted at 6 monthly intervals. Research blood samples will be collected at baseline, 6 and 12 months. Other correlative studies include matrix metalloproteinases (MMP), tissue inhibitor of metalloproteinases (TIMP) testing and patient-reported outcomes.
Collapse
|
45
|
Marcinko TM, Dong J, LeBlanc R, Daborowski KV, Vachet RW. Small molecule-mediated inhibition of β-2-microglobulin-based amyloid fibril formation. J Biol Chem 2017; 292:10630-10638. [PMID: 28468825 DOI: 10.1074/jbc.m116.774083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/02/2017] [Indexed: 12/26/2022] Open
Abstract
In dialysis patients, β-2 microglobulin (β2m) can aggregate and eventually form amyloid fibrils in a condition known as dialysis-related amyloidosis, which deleteriously affects joint and bone function. Recently, several small molecules have been identified as potential inhibitors of β2m amyloid formation in vitro Here we investigated whether these molecules are more broadly applicable inhibitors of β2m amyloid formation by studying their effect on Cu(II)-induced β2m amyloid formation. Using a variety of biophysical techniques, we also examined their inhibitory mechanisms. We found that two molecules, doxycycline and rifamycin SV, can inhibit β2m amyloid formation in vitro by causing the formation of amorphous, redissolvable aggregates. Rather than interfering with β2m amyloid formation at the monomer stage, we found that doxycycline and rifamycin SV exert their effect by binding to oligomeric species both in solution and in gas phase. Their binding results in a diversion of the expected Cu(II)-induced progression of oligomers toward a heterogeneous collection of oligomers, including trimers and pentamers, that ultimately matures into amorphous aggregates. Using ion mobility mass spectrometry, we show that both inhibitors promote the compaction of the initially formed β2m dimer, which causes the formation of other off-pathway and amyloid-incompetent oligomers that are isomeric with amyloid-competent oligomers in some cases. Overall, our results suggest that doxycycline and rifamycin are general inhibitors of Cu(II)-induced β2m amyloid formation. Interestingly, the putative mechanism of their activity is different depending on how amyloid formation is initiated with β2m, which underscores the complexity of how these structures assemble in vitro.
Collapse
Affiliation(s)
- Tyler M Marcinko
- From the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| | - Jia Dong
- From the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| | - Raquel LeBlanc
- From the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| | - Kate V Daborowski
- From the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| | - Richard W Vachet
- From the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| |
Collapse
|
46
|
Muchtar E, Gertz MA. Clinical trials evaluating potential therapies for light chain (AL) amyloidosis. Expert Opin Orphan Drugs 2017; 5:655-663. [PMID: 34567849 PMCID: PMC8460071 DOI: 10.1080/21678707.2017.1341834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The field of systemic amyloidosis is experiencing major advances in diagnostic and prognostic methods coupled with a growing availability in treatment options. AREAS COVERED Treatment of AL amyloidosis traditionally targeted the clonal plasma cells, in order to block further production of amyloidogenic light chains. Currently, a research focus is placed on targeting the already formed amyloid deposits using monoclonal antibodies against epitopes on such deposits. Encouraging results were obtained from the three investigated antibodies: NEOD001, 11-1F4 and anti-SAP, but further validation is required before these antibodies can be commercialized. In this paper, we review the current active clinical research in AL amyloidosis, which includes the monoclonal antibodies targeting amyloid deposits, daratumumab, Venetoclax, doxycycline, green tea, pomalidomide, carfilzomib and ixazomib. EXPERT OPINION Monoclonal antibodies, targeting either the amyloid deposits or the plasma cell compartment will likely be integrated into routine treatment practice given their encouraging results and minimal toxicity in the fragile population of AL amyloidosis. Other therapeutic options hold promise to the field as well, but toxicity will likely challenge their routine use. Early recognition remains the best option for outcome enhancement.
Collapse
Affiliation(s)
- Eli Muchtar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
47
|
Raimondi S, Porcari R, Mangione PP, Verona G, Marcoux J, Giorgetti S, Taylor GW, Ellmerich S, Ballico M, Zanini S, Pardon E, Al-Shawi R, Simons JP, Corazza A, Fogolari F, Leri M, Stefani M, Bucciantini M, Gillmore JD, Hawkins PN, Valli M, Stoppini M, Robinson CV, Steyaert J, Esposito G, Bellotti V. A specific nanobody prevents amyloidogenesis of D76N β 2-microglobulin in vitro and modifies its tissue distribution in vivo. Sci Rep 2017; 7:46711. [PMID: 28429761 PMCID: PMC5399440 DOI: 10.1038/srep46711] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/23/2017] [Indexed: 11/24/2022] Open
Abstract
Systemic amyloidosis is caused by misfolding and aggregation of globular proteins in vivo for which effective treatments are urgently needed. Inhibition of protein self-aggregation represents an attractive therapeutic strategy. Studies on the amyloidogenic variant of β2-microglobulin, D76N, causing hereditary systemic amyloidosis, have become particularly relevant since fibrils are formed in vitro in physiologically relevant conditions. Here we compare the potency of two previously described inhibitors of wild type β2-microglobulin fibrillogenesis, doxycycline and single domain antibodies (nanobodies). The β2-microglobulin -binding nanobody, Nb24, more potently inhibits D76N β2-microglobulin fibrillogenesis than doxycycline with complete abrogation of fibril formation. In β2-microglobulin knock out mice, the D76N β2-microglobulin/ Nb24 pre-formed complex, is cleared from the circulation at the same rate as the uncomplexed protein; however, the analysis of tissue distribution reveals that the interaction with the antibody reduces the concentration of the variant protein in the heart but does not modify the tissue distribution of wild type β2-microglobulin. These findings strongly support the potential therapeutic use of this antibody in the treatment of systemic amyloidosis.
Collapse
Affiliation(s)
- Sara Raimondi
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy
| | - Riccardo Porcari
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - P Patrizia Mangione
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy.,Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Guglielmo Verona
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Julien Marcoux
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Sofia Giorgetti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy
| | - Graham W Taylor
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Stephan Ellmerich
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Maurizio Ballico
- Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, UAE
| | - Stefano Zanini
- Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, UAE
| | - Els Pardon
- Structural Biology Research Centre, VIB, Pleinlaan 2, 1050, Brussel, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussel, Belgium
| | - Raya Al-Shawi
- Centre for Biomedical Science, Division of Medicine, University College London, London NW3 2PF, UK
| | - J Paul Simons
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Alessandra Corazza
- Department of Medical and Biological Sciences (DSMB), University of Udine, Piazzale Kolbe 4, 33100 Udine, Italy.,Istituto Nazionale Biostrutture e Biosistemi, Viale Medaglie d'Oro 305, 00136 Roma, Italy
| | - Federico Fogolari
- Istituto Nazionale Biostrutture e Biosistemi, Viale Medaglie d'Oro 305, 00136 Roma, Italy.,Department of Mathematics, Computer Science and Physics, University of Udine, Piazzale Kolbe 4, 33100 Udine, Italy
| | - Manuela Leri
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Massimo Stefani
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Viale Morgagni 50, 50134 Florence, Italy.,Research Centre for Molecular Basis of Neurodegeneration, 50134 Florence, Italy
| | - Monica Bucciantini
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Viale Morgagni 50, 50134 Florence, Italy.,Research Centre for Molecular Basis of Neurodegeneration, 50134 Florence, Italy
| | - Julian D Gillmore
- National Amyloidosis Centre, University College London, London NW3 2PF, UK
| | - Philip N Hawkins
- National Amyloidosis Centre, University College London, London NW3 2PF, UK
| | - Maurizia Valli
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy
| | - Monica Stoppini
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy
| | - Carol V Robinson
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Jan Steyaert
- Structural Biology Research Centre, VIB, Pleinlaan 2, 1050, Brussel, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussel, Belgium
| | - Gennaro Esposito
- Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, UAE.,Istituto Nazionale Biostrutture e Biosistemi, Viale Medaglie d'Oro 305, 00136 Roma, Italy.,Department of Mathematics, Computer Science and Physics, University of Udine, Piazzale Kolbe 4, 33100 Udine, Italy
| | - Vittorio Bellotti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Via Taramelli 3b, 27100 Pavia, Italy.,Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| |
Collapse
|
48
|
The amyloidogenic light chain is a stressor that sensitizes plasma cells to proteasome inhibitor toxicity. Blood 2017; 129:2132-2142. [DOI: 10.1182/blood-2016-08-730978] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 01/09/2017] [Indexed: 01/22/2023] Open
Abstract
Key PointsAmyloidogenic PCs show unique PI susceptibility and altered organelle homeostasis, consistent with defective autophagy. Amyloidogenic LC production is an intrinsic cellular stressor that sensitizes to PI toxicity.
Collapse
|
49
|
Cantarutti C, Raimondi S, Brancolini G, Corazza A, Giorgetti S, Ballico M, Zanini S, Palmisano G, Bertoncin P, Marchese L, Patrizia Mangione P, Bellotti V, Corni S, Fogolari F, Esposito G. Citrate-stabilized gold nanoparticles hinder fibrillogenesis of a pathological variant of β 2-microglobulin. NANOSCALE 2017; 9:3941-3951. [PMID: 28265615 DOI: 10.1039/c6nr09362k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Nanoparticles have repeatedly been shown to enhance fibril formation when assayed with amyloidogenic proteins. Recently, however, evidence casting some doubt about the generality of this conclusion started to emerge. Therefore, to investigate further the influence of nanoparticles on the fibrillation process, we used a naturally occurring variant of the paradigmatic amyloidogenic protein β2-microglobulin (β2m), namely D76N β2m where asparagine replaces aspartate at position 76. This variant is responsible for aggressive systemic amyloidosis. After characterizing the interaction of the variant with citrate-stabilized gold nanoparticles (Cit-AuNPs) by NMR and modeling, we analyzed the fibril formation by three different methods: thioflavin T fluorescence, native agarose gel electrophoresis and transmission electron microscopy. The NMR evidence indicated a fast-exchange interaction involving preferentially specific regions of the protein that proved, by subsequent modeling, to be consistent with a dimeric adduct interacting with Cit-AuNPs. The fibril detection assays showed that AuNPs are able to hamper D76N β2m fibrillogenesis through an effective interaction that competes with protofibril formation or recruitment. These findings open promising perspectives for the optimization of the nanoparticle surface to design tunable interactions with proteins.
Collapse
Affiliation(s)
| | - Sara Raimondi
- Dipartimento Medicina Molecolare, Università di Pavia, Via Taramelli 3, 27100 Pavia, Italy and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy
| | | | - Alessandra Corazza
- DSMB, Università di Udine, P.le Kolbe 4, 33100 Udine, Italy. and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy
| | - Sofia Giorgetti
- Dipartimento Medicina Molecolare, Università di Pavia, Via Taramelli 3, 27100 Pavia, Italy and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy
| | - Maurizio Ballico
- Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Stefano Zanini
- Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Giovanni Palmisano
- Department of Chemical and Environmental Engineering, Masdar Institute of Science and Technology, PO Box 54224, Abu Dhabi, United Arab Emirates
| | - Paolo Bertoncin
- Dipartimento Scienze della Vita, Università di Trieste, Via Weiss 2, 34128 Trieste, Italy
| | - Loredana Marchese
- Dipartimento Medicina Molecolare, Università di Pavia, Via Taramelli 3, 27100 Pavia, Italy and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy
| | - P Patrizia Mangione
- Dipartimento Medicina Molecolare, Università di Pavia, Via Taramelli 3, 27100 Pavia, Italy and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy and Division of Medicine, University College of London, London NW3 2PF, UK
| | - Vittorio Bellotti
- Dipartimento Medicina Molecolare, Università di Pavia, Via Taramelli 3, 27100 Pavia, Italy and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy and Division of Medicine, University College of London, London NW3 2PF, UK
| | - Stefano Corni
- CNR Istituto Nanoscienze, Via Campi 213/A, 41125 Modena, Italy.
| | - Federico Fogolari
- DSMB, Università di Udine, P.le Kolbe 4, 33100 Udine, Italy. and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy
| | - Gennaro Esposito
- DSMB, Università di Udine, P.le Kolbe 4, 33100 Udine, Italy. and INBB, Viale Medaglie d'Oro 305, 00136 Roma, Italy and Science and Math Division, New York University at Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
50
|
González-Lizárraga F, Socías SB, Ávila CL, Torres-Bugeau CM, Barbosa LRS, Binolfi A, Sepúlveda-Díaz JE, Del-Bel E, Fernandez CO, Papy-Garcia D, Itri R, Raisman-Vozari R, Chehín RN. Repurposing doxycycline for synucleinopathies: remodelling of α-synuclein oligomers towards non-toxic parallel beta-sheet structured species. Sci Rep 2017; 7:41755. [PMID: 28155912 PMCID: PMC5290535 DOI: 10.1038/srep41755] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/14/2016] [Indexed: 12/03/2022] Open
Abstract
Synucleinophaties are progressive neurodegenerative disorders with no cure to date. An attractive strategy to tackle this problem is repurposing already tested safe drugs against novel targets. In this way, doxycycline prevents neurodegeneration in Parkinson models by modulating neuroinflammation. However, anti-inflammatory therapy per se is insufficient to account for neuroprotection. Herein we characterise novel targets of doxycycline describing the structural background supporting its effectiveness as a neuroprotector at subantibiotic doses. Our results show that doxycycline reshapes α-synuclein oligomers into off-pathway, high-molecular-weight species that do not evolve into fibrils. Off-pathway species present less hydrophobic surface than on-pathway oligomers and display different β-sheet structural arrangement. These structural changes affect the α-synuclein ability to destabilize biological membranes, cell viability, and formation of additional toxic species. Altogether, these mechanisms could act synergically giving novel targets for repurposing this drug.
Collapse
Affiliation(s)
- Florencia González-Lizárraga
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CCT-Tucumán and Instituto de Química Biológica Dr Bernabé Bloj (CONICET-UNT), Chacabuco 461 (T4000ILI) Tucumán, Argentina.,Institut National De La Santé Et De La Recherche Médicale, U 1127, CNRS, Unité Mixte De Recherche (UMR) 7225, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Institut Du Cerveau Et De La Moelle Epinière, ICM, Paris, France
| | - Sergio B Socías
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CCT-Tucumán and Instituto de Química Biológica Dr Bernabé Bloj (CONICET-UNT), Chacabuco 461 (T4000ILI) Tucumán, Argentina
| | - César L Ávila
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CCT-Tucumán and Instituto de Química Biológica Dr Bernabé Bloj (CONICET-UNT), Chacabuco 461 (T4000ILI) Tucumán, Argentina
| | - Clarisa M Torres-Bugeau
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CCT-Tucumán and Instituto de Química Biológica Dr Bernabé Bloj (CONICET-UNT), Chacabuco 461 (T4000ILI) Tucumán, Argentina
| | - Leandro R S Barbosa
- Instituto de Física da Universidade de São Paulo - IFUSP, Rua do Matão, Travessa R, 187, São Paulo, Brazil
| | - Andres Binolfi
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC) and Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario, Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Julia E Sepúlveda-Díaz
- Institut National De La Santé Et De La Recherche Médicale, U 1127, CNRS, Unité Mixte De Recherche (UMR) 7225, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Institut Du Cerveau Et De La Moelle Epinière, ICM, Paris, France
| | - Elaine Del-Bel
- Department of Morphology, Physiology and Stomatology, Faculty of Odontology of Ribeirão Preto, University of São Paulo, Brazil, Center of Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, Brazil
| | - Claudio O Fernandez
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC) and Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario, Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Dulce Papy-Garcia
- Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), CNRS ERL 9215, Université Paris Est Créteil, Université Paris Est, F-94000, Créteil, France
| | - Rosangela Itri
- Instituto de Física da Universidade de São Paulo - IFUSP, Rua do Matão, Travessa R, 187, São Paulo, Brazil
| | - Rita Raisman-Vozari
- Institut National De La Santé Et De La Recherche Médicale, U 1127, CNRS, Unité Mixte De Recherche (UMR) 7225, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Institut Du Cerveau Et De La Moelle Epinière, ICM, Paris, France
| | - Rosana N Chehín
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CCT-Tucumán and Instituto de Química Biológica Dr Bernabé Bloj (CONICET-UNT), Chacabuco 461 (T4000ILI) Tucumán, Argentina
| |
Collapse
|