1
|
Tsekrekou M, Giannakou M, Papanikolopoulou K, Skretas G. Protein aggregation and therapeutic strategies in SOD1- and TDP-43- linked ALS. Front Mol Biosci 2024; 11:1383453. [PMID: 38855322 PMCID: PMC11157337 DOI: 10.3389/fmolb.2024.1383453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 06/11/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with severe socio-economic impact. A hallmark of ALS pathology is the presence of aberrant cytoplasmic inclusions composed of misfolded and aggregated proteins, including both wild-type and mutant forms. This review highlights the critical role of misfolded protein species in ALS pathogenesis, particularly focusing on Cu/Zn superoxide dismutase (SOD1) and TAR DNA-binding protein 43 (TDP-43), and emphasizes the urgent need for innovative therapeutic strategies targeting these misfolded proteins directly. Despite significant advancements in understanding ALS mechanisms, the disease remains incurable, with current treatments offering limited clinical benefits. Through a comprehensive analysis, the review focuses on the direct modulation of the misfolded proteins and presents recent discoveries in small molecules and peptides that inhibit SOD1 and TDP-43 aggregation, underscoring their potential as effective treatments to modify disease progression and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Tsekrekou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Maria Giannakou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Papanikolopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece
- ResQ Biotech, Patras Science Park, Rio, Greece
| | - Georgios Skretas
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- ResQ Biotech, Patras Science Park, Rio, Greece
- Institute for Bio-innovation, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece
| |
Collapse
|
2
|
Elmansy MF, Reidl CT, Rahaman M, Özdinler PH, Silverman RB. Small molecules targeting different cellular pathologies for the treatment of amyotrophic lateral sclerosis. Med Res Rev 2023; 43:2260-2302. [PMID: 37243319 PMCID: PMC10592673 DOI: 10.1002/med.21974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 02/28/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease in which the motor neuron circuitry displays progressive degeneration, affecting mostly the motor neurons in the brain and in the spinal cord. There are no effective cures, albeit three drugs, riluzole, edaravone, and AMX0035 (a combination of sodium phenylbutyrate and taurursodiol), have been approved by the Food and Drug Administration, with limited improvement in patients. There is an urgent need to build better and more effective treatment strategies for ALS. Since the disease is very heterogenous, numerous approaches have been explored, such as targeting genetic mutations, decreasing oxidative stress and excitotoxicity, enhancing mitochondrial function and protein degradation mechanisms, and inhibiting neuroinflammation. In addition, various chemical libraries or previously identified drugs have been screened for potential repurposing in the treatment of ALS. Here, we review previous drug discovery efforts targeting a variety of cellular pathologies that occur from genetic mutations that cause ALS, such as mutations in SOD1, C9orf72, FUS, and TARDP-43 genes. These mutations result in protein aggregation, which causes neuronal degeneration. Compounds used to target cellular pathologies that stem from these mutations are discussed and comparisons among different preclinical models are presented. Because the drug discovery landscape for ALS and other motor neuron diseases is changing rapidly, we also offer recommendations for a novel, more effective, direction in ALS drug discovery that could accelerate translation of effective compounds from animals to patients.
Collapse
Affiliation(s)
- Mohamed F. Elmansy
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
- Department of Organometallic and Organometalloid Chemistry, National Research Centre, Cairo, Egypt
| | - Cory T. Reidl
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
| | - Mizzanoor Rahaman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
| | - P. Hande Özdinler
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
3
|
Gupta A, Laha JK. Growing Utilization of Radical Chemistry in the Synthesis of Pharmaceuticals. CHEM REC 2023; 23:e202300207. [PMID: 37565381 DOI: 10.1002/tcr.202300207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/19/2023] [Indexed: 08/12/2023]
Abstract
Our current unhealthy lifestyle and the exponential surge in the population getting affected by a variety of diseases have made pharmaceuticals or drugs an imperative part of life, making the development of innovative strategies for drug discovery or the introduction of refined, cost-effective and modern technologies for the synthesis of clinically used drugs, a need of the hour. Ever since their discovery, free radicals and radical cations or anions as reactive intermediates have captivated the chemists, resulting in an exceptional utilization of these moieties throughout the field of chemical synthesis, owing to their unprecedented and widespread reactivity. Sticking with the idea of not judging the book by its cover, despite the conventional thought process of radicals being unstable and difficult to control entities, scientists and academicians around the globe have done an appreciable amount of work utilizing both persistent as well as transient radicals for a variety of organic transformations, exemplifying them with the synthesis of significant biologically active pharmaceutical ingredients. This review truly accounts for the organic radical transformations including radical addition, radical cascade cyclization, radical/radical cross-coupling, coupling with metal-complexes and radical cations coupling with nucleophiles, that offers fascinating and unconventional approaches towards the construction of intricate structural frameworks of marketed APIs with high atom- and step-economy; complementing the otherwise employed traditional methods. This tutorial review presents a comprehensive package of diverse methods utilized for radical generation, featuring their reactivity to form critical bonds in pharmaceutical total synthesis or in building key starting materials or intermediates of their synthetic journey, acknowledging their excellence, downsides and underlying mechanisms, which are otherwise poorly highlighted in the literature. Despite great achievements over the past few decades in this area, many challenges and obstacles are yet to be unraveled to shorten the distance between the academics and the industry, which are all discussed in summary and outlook.
Collapse
Affiliation(s)
- Anjali Gupta
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education & Research (NIPER) S.A.S. Nagar, Sahibzada Ajit Singh Nagar, Mohali, 160062, India
| | - Joydev K Laha
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education & Research (NIPER) S.A.S. Nagar, Sahibzada Ajit Singh Nagar, Mohali, 160062, India
| |
Collapse
|
4
|
Spennato M, Roggero OM, Varriale S, Asaro F, Cortesi A, Kašpar J, Tongiorgi E, Pezzella C, Gardossi L. Neuroprotective Properties of Cardoon Leaves Extracts against Neurodevelopmental Deficits in an In Vitro Model of Rett Syndrome Depend on the Extraction Method and Harvest Time. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248772. [PMID: 36557905 PMCID: PMC9783035 DOI: 10.3390/molecules27248772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
This study investigates the bioactive properties of different extracts of cardoon leaves in rescuing neuronal development arrest in an in vitro model of Rett syndrome (RTT). Samples were obtained from plants harvested at different maturity stages and extracted with two different methodologies, namely Naviglio® and supercritical carbon dioxide (scCO2). While scCO2 extracts more hydrophobic fractions, the Naviglio® method extracts phenolic compounds and less hydrophobic components. Only the scCO2 cardoon leaves extract obtained from plants harvested in spring induced a significant rescue of neuronal atrophy in RTT neurons, while the scCO2 extract from the autumn harvest stimulated dendrite outgrowth in Wild-Type (WT) neurons. The scCO2 extracts were the richest in squalene, 3ß-taraxerol and lupeol, with concentrations in autumn harvest doubling those in spring harvest. The Naviglio® extract was rich in cynaropicrin and exerted a toxic effect at 20 µM on both WT and RTT neurons. When cynaropicrin, squalene, lupeol and 3ß-taraxerol were tested individually, no positive effect was observed, whereas a significant neurotoxicity of cynaropicrin and lupeol was evident. In conclusion, cardoon leaves extracts with high content of hydrophobic bioactive molecules and low cynaropicrin and lupeol concentrations have pharmacological potential to stimulate neuronal development in RTT and WT neurons in vitro.
Collapse
Affiliation(s)
- Mariachiara Spennato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Ottavia Maria Roggero
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 5, 34127 Trieste, Italy
| | - Simona Varriale
- Department of Chemical Sciences, University Federico II of Naples, Via Cinthia, 4, 80126 Napoli, Italy
| | - Fioretta Asaro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Angelo Cortesi
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio 6/A, 34127 Trieste, Italy
| | - Jan Kašpar
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 5, 34127 Trieste, Italy
| | - Cinzia Pezzella
- Department of Chemical Sciences, University Federico II of Naples, Via Cinthia, 4, 80126 Napoli, Italy
| | - Lucia Gardossi
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
- Correspondence:
| |
Collapse
|
5
|
Pampalakis G, Angelis G, Zingkou E, Vekrellis K, Sotiropoulou G. A chemogenomic approach is required for effective treatment of amyotrophic lateral sclerosis. Clin Transl Med 2022; 12:e657. [PMID: 35064780 PMCID: PMC8783349 DOI: 10.1002/ctm2.657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 11/10/2022] Open
Abstract
ALS is a fatal untreatable disease involving degeneration of motor neurons. Μultiple causative genes encoding proteins with versatile functions have been identified indicating that diverse biological pathways lead to ALS. Chemical entities still represent a promising choice to delay ALS progression, attenuate symptoms and/or increase life expectancy, but also gene-based and stem cell-based therapies are in the process of development, and some are tested in clinical trials. Various compounds proved effective in transgenic models overexpressing distinct ALS causative genes unfortunately though, they showed no efficacy in clinical trials. Notably, while animal models provide a uniform genetic background for preclinical testing, ALS patients are not stratified, and the distinct genetic forms of ALS are treated as one group, which could explain the observed discrepancies between treating genetically homogeneous mice and quite heterogeneous patient cohorts. We suggest that chemical entity-genotype correlation should be exploited to guide patient stratification for pharmacotherapy, that is administered drugs should be selected based on the ALS genetic background.
Collapse
Affiliation(s)
- Georgios Pampalakis
- Department of Pharmacology - Pharmacognosy, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Angelis
- Department of Pharmacology - Pharmacognosy, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Kostas Vekrellis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| |
Collapse
|
6
|
Strategies in the design and development of (TAR) DNA-binding protein 43 (TDP-43) binding ligands. Eur J Med Chem 2021; 225:113753. [PMID: 34388383 DOI: 10.1016/j.ejmech.2021.113753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 01/09/2023]
Abstract
The human transactive responsive (TAR) DNA-binding protein 43 (TDP-43) is involved in a number of physiological processes in the body. Its primary function involves RNA regulation. The TDP-43 protein is also involved in many diseases such as amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease (AD), Parkinson's disease (PD) and even cancers. These TDP-43 mediated diseases are collectively called as TDP-43 proteinopathies. Intense research in the last decade has increased our understanding on TDP-43 structure and function in biology. The three-dimensional structures of TDP-43 domains such as N-terminal domain (NTD), RNA-recognition motif-1 (RRM1), RNA-recognition motif-2 (RRM2) and the C-terminal domain (CTD) or low-complexity domain (LCD) have been solved. These structures have yielded insights into novel binding sites and pockets at various TDP-43 domains, which can be targeted by designing a diverse library of ligands including small molecules, peptides and oligonucleotides as molecular tools to (i) study TDP-43 function, (ii) develop novel diagnostic agents and (iii) discover disease-modifying therapies to treat TDP-43 proteinopathies. This review provides a summary on recent progress in the development of TDP-43 binding ligands and uses the solved structures of various TDP-43 domains to investigate putative ligand binding regions that can be exploited to discover novel molecular probes to modulate TDP-43 structure and function.
Collapse
|
7
|
Genç B, Gautam M, Gözütok Ö, Dervishi I, Sanchez S, Goshu GM, Koçak N, Xie E, Silverman RB, Özdinler PH. Improving mitochondria and ER stability helps eliminate upper motor neuron degeneration that occurs due to mSOD1 toxicity and TDP-43 pathology. Clin Transl Med 2021; 11:e336. [PMID: 33634973 PMCID: PMC7898037 DOI: 10.1002/ctm2.336] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Upper motor neurons (UMNs) are a key component of motor neuron circuitry. Their degeneration is a hallmark for diseases, such as hereditary spastic paraplegia (HSP), primary lateral sclerosis (PLS), and amyotrophic lateral sclerosis (ALS). Currently there are no preclinical assays investigating cellular responses of UMNs to compound treatment, even for diseases of the UMNs. The basis of UMN vulnerability is not fully understood, and no compound has yet been identified to improve the health of diseased UMNs: two major roadblocks for building effective treatment strategies. METHODS Novel UMN reporter models, in which UMNs that are diseased because of misfolded superoxide dismutase protein (mSOD1) toxicity and TDP-43 pathology are labeled with eGFP expression, allow direct assessment of UMN response to compound treatment. Electron microscopy reveals very precise aspects of endoplasmic reticulum (ER) and mitochondrial damage. Administration of NU-9, a compound initially identified based on its ability to reduce mSOD1 toxicity, has profound impact on improving the health and stability of UMNs, as identified by detailed cellular and ultrastructural analyses. RESULTS Problems with mitochondria and ER are conserved in diseased UMNs among different species. NU-9 has drug-like pharmacokinetic properties. It lacks toxicity and crosses the blood brain barrier. NU-9 improves the structural integrity of mitochondria and ER, reduces levels of mSOD1, stabilizes degenerating UMN apical dendrites, improves motor behavior measured by the hanging wire test, and eliminates ongoing degeneration of UMNs that become diseased both because of mSOD1 toxicity and TDP-43 pathology, two distinct and important overarching causes of motor neuron degeneration. CONCLUSIONS Mechanism-focused and cell-based drug discovery approaches not only addressed key cellular defects responsible for UMN loss, but also identified NU-9, the first compound to improve the health of diseased UMNs, neurons that degenerate in ALS, HSP, PLS, and ALS/FTLD patients.
Collapse
Affiliation(s)
- Barış Genç
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Mukesh Gautam
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Öge Gözütok
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Ina Dervishi
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Santana Sanchez
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Gashaw M. Goshu
- Department of ChemistryNorthwestern UniversityEvanstonIllinoisUSA
| | - Nuran Koçak
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Edward Xie
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Richard B. Silverman
- Department of ChemistryNorthwestern UniversityEvanstonIllinoisUSA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental TherapeuticsNorthwestern UniversityEvanstonIllinoisUSA
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIL60208
| | - P. Hande Özdinler
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental TherapeuticsNorthwestern UniversityEvanstonIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIL60208
- Mesulam Center for Cognitive Neurology and Alzheimer's DiseaseNorthwestern University, Feinberg School of MedicineChicagoIL60611
- Les Turner ALS CenterNorthwestern University, Feinberg School of MedicineChicagoIL60611
| |
Collapse
|
8
|
Wang G, Rayner S, Chung R, Shi B, Liang X. Advances in nanotechnology-based strategies for the treatments of amyotrophic lateral sclerosis. Mater Today Bio 2020; 6:100055. [PMID: 32529183 PMCID: PMC7280770 DOI: 10.1016/j.mtbio.2020.100055] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/09/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease (MND), is a progressive neurodegenerative disease that affects both upper and lower motor neurons, which results in loss of muscle control and eventual paralysis [1]. Currently, there are as yet unresolved challenges regarding efficient drug delivery into the central nervous system (CNS). These challenges can be attributed to multiple factors including the presence of the blood-brain barrier (BBB), blood-spinal cord barrier (BSCB), as well as the inherent characteristics of the drugs themselves (e.g. low solubility, insufficient bioavailability/bio-stability, 'off-target' effects) etc. As a result, conventional drug delivery systems may not facilitate adequate dosage of the required drugs for functional recovery in ALS patients. Nanotechnology-based strategies, however, employ engineered nanostructures that show great potential in delivering single or combined therapeutic agents to overcome the biological barriers, enhance interaction with targeted sites, improve drug bioavailability/bio-stability and achieve real-time tracking while minimizing the systemic side-effects. This review provides a concise discussion of recent advances in nanotechnology-based strategies in relation to combating specific pathophysiology relevant to ALS disease progression and investigates the future scope of using nanotechnology to develop innovative treatments for ALS patients.
Collapse
Affiliation(s)
- G.Y. Wang
- Huaihe Hospital, Henan University, Kaifeng, Henan, 475004, China
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - S.L. Rayner
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - R. Chung
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - B.Y. Shi
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - X.J. Liang
- Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
9
|
Sharma V, Ghosh KS. Inhibition of Amyloid Fibrillation by Small Molecules and Nanomaterials: Strategic Development of Pharmaceuticals Against Amyloidosis. Protein Pept Lett 2019; 26:315-323. [PMID: 30848182 DOI: 10.2174/0929866526666190307164944] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
Amyloid fibrils are a special class of self-assembled protein molecules, which exhibit various toxic effects in cells. Different physiological disorders such as Alzheimer's, Parkinson's, Huntington's diseases, etc. happen due to amyloid formation and lack of proper cellular mechanism for the removal of fibrils. Therefore, inhibition of amyloid fibrillation will find immense applications to combat the diseases associated with amyloidosis. The development of therapeutics against amyloidosis is definitely challenging and numerous strategies have been followed to find out anti-amyloidogenic molecules. Inhibition of amyloid aggregation of proteins can be achieved either by stabilizing the native conformation or by decreasing the chances of assembly formation by the unfolded/misfolded structures. Various small molecules such as naturally occurring polyphenols, flavonoids, small organic molecules, surfactants, dyes, chaperones, etc. have demonstrated their capability to interrupt the amyloid fibrillation of proteins. In addition to that, in last few years, different nanomaterials were evolved as effective therapeutic inhibitors against amyloidosis. Aromatic and hydrophobic interactions between the partially unfolded protein molecules and the inhibitors had been pointed as a general mechanism for inhibition. In this review article, we are presenting an overview on the inhibition of amyloidosis by using different small molecules (both natural and synthetic origin) as well as nanomaterials for development of pharmaceutical strategies against amyloid diseases.
Collapse
Affiliation(s)
- Vandna Sharma
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh 177005, India
| | - Kalyan Sundar Ghosh
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh 177005, India
| |
Collapse
|
10
|
Zhu Y, Li Y, Xiang S, Fan W, Jin J, Huang D. Utilization of nitriles as the nitrogen source: practical and economical construction of 4-aminopyrimidine and β-enaminonitrile skeletons. Org Chem Front 2019. [DOI: 10.1039/c9qo00619b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A highly practical and economical method for the synthesis of 4-aminopyrimidines and β-enaminonitriles from mixed organonitriles is reported.
Collapse
Affiliation(s)
- Yingzu Zhu
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou
- China
| | - Yinghua Li
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou
- China
| | - Shiqun Xiang
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou
- China
| | - Weibin Fan
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou
- China
| | - Jiang Jin
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou
- China
| | - Deguang Huang
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou
- China
| |
Collapse
|
11
|
Ramalho TC, de Castro AA, Tavares TS, Silva MC, Silva DR, Cesar PH, Santos LA, da Cunha EFF, Nepovimova E, Kuca K. Insights into the pharmaceuticals and mechanisms of neurological orphan diseases: Current Status and future expectations. Prog Neurobiol 2018; 169:135-157. [PMID: 29981392 DOI: 10.1016/j.pneurobio.2018.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 06/30/2018] [Indexed: 12/20/2022]
Abstract
Several rare or orphan diseases have been characterized that singly affect low numbers of people, but cumulatively reach ∼6%-10% of the population in Europe and in the United States. Human genetics has shown to be broadly effective when evaluating subjacent genetic defects such as orphan genetic diseases, but on the other hand, a modest progress has been achieved toward comprehending the molecular pathologies and designing new therapies. Chemical genetics, placed at the interface of chemistry and genetics, could be employed to understand the molecular mechanisms of subjacent illnesses and for the discovery of new remediation processes. This review debates current progress in chemical genetics, and how a variety of compounds and reaction mechanisms can be used to study and ultimately treat rare genetic diseases. We focus here on a study involving Amyotrophic lateral sclerosis (ALS), Duchenne Muscular Dystrophy (DMD), Spinal muscular atrophy (SMA) and Familial Amyloid Polyneuropathy (FAP), approaching different treatment methods and the reaction mechanisms of several compounds, trying to elucidate new routes capable of assisting in the treatment profile.
Collapse
Affiliation(s)
- Teodorico C Ramalho
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil; Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| | | | - Tássia S Tavares
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Maria C Silva
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Daniela R Silva
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Pedro H Cesar
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Lucas A Santos
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Elaine F F da Cunha
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
12
|
Heterogeneous gold(I)-catalyzed [2 + 2 + 2] annulation between ynamides and nitriles: Straightforward synthesis of tetrasubstituted pyrimidines. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
13
|
Kim JM, Billington E, Reyes A, Notarianni T, Sage J, Agbas E, Taylor M, Monast I, Stanford JA, Agbas A. Impaired Cu-Zn Superoxide Dismutase (SOD1) and Calcineurin (Cn) Interaction in ALS: A Presumed Consequence for TDP-43 and Zinc Aggregation in Tg SOD1 G93A Rodent Spinal Cord Tissue. Neurochem Res 2018; 44:228-233. [PMID: 29299811 PMCID: PMC6345727 DOI: 10.1007/s11064-017-2461-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
Impaired interactions between Calcineurin (Cn) and (Cu/Zn) superoxide dismutase (SOD1) are suspected to be responsible for the formation of hyperphosphorylated protein aggregation in amyotrophic lateral sclerosis (ALS). Serine (Ser)- enriched phosphorylated TDP-43 protein aggregation appears in the spinal cord of ALS animal models, and may be linked to the reduced phosphatase activity of Cn. The mutant overexpressed SOD1G93A protein does not properly bind zinc (Zn) in animal models; hence, mutant SOD1G93A–Cn interaction weakens. Consequently, unstable Cn fails to dephosphorylate TDP-43 that yields hyperphosphorylated TDP-43 aggregates. Our previous studies had suggested that Cn and SOD1 interaction was necessary to keep Cn enzyme functional. We have observed low Cn level, increased Zn concentrations, and increased TDP-43 protein levels in cervical, thoracic, lumbar, and sacral regions of the spinal cord tissue homogenates. This study further supports our previously published work indicating that Cn stability depends on functional Cn–SOD1 interaction because Zn is crucial for maintaining the Cn stability. Less active Cn did not efficiently dephosphorylate TDP-43; hence TDP-43 aggregations appeared in the spinal cord tissue.
Collapse
Affiliation(s)
- Jolene M Kim
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - Elizabeth Billington
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - Ada Reyes
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - Tara Notarianni
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - Jessica Sage
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - Emre Agbas
- Stowers Institute for Medical Research, 1000 E 50th Street, Kansas City, MO, 64110, USA
| | - Michael Taylor
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - Ian Monast
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA
| | - John A Stanford
- University of Kansas Medical Center, 3901 Rainbow Blvd., 2096 HLSIC, Kansas City, KS, 66160, USA
| | - Abdulbaki Agbas
- Division of Basic Sciences, College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, 1750 Independence Avenue, Kansas City, MO, 6410, USA.
| |
Collapse
|
14
|
Can Astrocytes Be a Target for Precision Medicine? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:111-128. [DOI: 10.1007/978-3-319-60733-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Banerjee V, Oren O, Ben-Zeev E, Taube R, Engel S, Papo N. A computational combinatorial approach identifies a protein inhibitor of superoxide dismutase 1 misfolding, aggregation, and cytotoxicity. J Biol Chem 2017; 292:15777-15788. [PMID: 28768772 DOI: 10.1074/jbc.m117.789610] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/21/2017] [Indexed: 12/12/2022] Open
Abstract
Molecular agents that specifically bind and neutralize misfolded and toxic superoxide dismutase 1 (SOD1) mutant proteins may find application in attenuating the disease progression of familial amyotrophic lateral sclerosis. However, high structural similarities between the wild-type and mutant SOD1 proteins limit the utility of this approach. Here we addressed this challenge by converting a promiscuous natural human IgG-binding domain, the hyperthermophilic variant of protein G (HTB1), into a highly specific aggregation inhibitor (designated HTB1M) of two familial amyotrophic lateral sclerosis-linked SOD1 mutants, SOD1G93A and SOD1G85R We utilized a computational algorithm for mapping protein surfaces predisposed to HTB1 intermolecular interactions to construct a focused HTB1 library, complemented with an experimental platform based on yeast surface display for affinity and specificity screening. HTB1M displayed high binding specificity toward SOD1 mutants, inhibited their amyloid aggregation in vitro, prevented the accumulation of misfolded proteins in living cells, and reduced the cytotoxicity of SOD1G93A expressed in motor neuron-like cells. Competition assays and molecular docking simulations suggested that HTB1M binds to SOD1 via both its α-helical and β-sheet domains at the native dimer interface that becomes exposed upon mutated SOD1 misfolding and monomerization. Our results demonstrate the utility of computational mapping of the protein-protein interaction potential for designing focused protein libraries to be used in directed evolution. They also provide new insight into the mechanism of conversion of broad-spectrum immunoglobulin-binding proteins, such as HTB1, into target-specific proteins, thereby paving the way for the development of new selective drugs targeting the amyloidogenic proteins implicated in a variety of human diseases.
Collapse
Affiliation(s)
- Victor Banerjee
- From the Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ofek Oren
- From the Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.,the Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel, and
| | - Efrat Ben-Zeev
- the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovoth 76100, Israel
| | - Ran Taube
- the Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel, and
| | - Stanislav Engel
- the Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Niv Papo
- From the Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel,
| |
Collapse
|
16
|
Kreiner G, Rafa-Zabłocka K, Chmielarz P, Bagińska M, Nalepa I. Lack of riluzole efficacy in the progression of the neurodegenerative phenotype in a new conditional mouse model of striatal degeneration. PeerJ 2017; 5:e3240. [PMID: 28462043 PMCID: PMC5410142 DOI: 10.7717/peerj.3240] [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: 10/28/2016] [Accepted: 03/28/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is a rare familial autosomal dominant neurodegenerative disorder characterized by progressive degeneration of medium spiny neurons (MSNs) located in the striatum. Currently available treatments of HD are only limited to alleviating symptoms; therefore, high expectations for an effective therapy are associated with potential replacement of lost neurons through stimulation of postnatal neurogenesis. One of the drugs of potential interest for the treatment of HD is riluzole, which may act as a positive modulator of adult neurogenesis, promoting replacement of damaged MSNs. The aim of this study was to evaluate the effects of chronic riluzole treatment on a novel HD-like transgenic mouse model, based on the genetic ablation of the transcription factor TIF-IA. This model is characterized by selective and progressive degeneration of MSNs. METHODS Selective ablation of TIF-IA in MSNs (TIF-IAD1RCre mice) was achieved by Cre-based recombination driven by the dopamine 1 receptor (D1R) promoter in the C57Bl/6N mouse strain. Riluzole was administered for 14 consecutive days (5 mg/kg, i.p.; 1× daily) starting at six weeks of age. Behavioral analysis included a motor coordination test performed on 13-week-old animals on an accelerated rotarod (4-40 r.p.m.; 5 min). To visualize the potential effects of riluzole treatment, the striata of the animals were stained by immunohistochemistry (IHC) and/or immunofluorescence (IF) with Ki67 (marker of proliferating cells), neuronal markers (NeuN, MAP2, DCX), and markers associated with neurodegeneration (GFAP, 8OHdG, FluoroJade C). Additionally, the morphology of dendritic spines of neurons was assessed by a commercially available FD Rapid Golgi Stain™ Kit. RESULTS A comparative analysis of IHC staining patterns with chosen markers for the neurodegeneration process in MSNs did not show an effect of riluzole on delaying the progression of MSN cell death despite an observed enhancement of cell proliferation as visualized by the Ki67 marker. A lack of a riluzole effect was also reflected by the behavioral phenotype associated with MSN degeneration. Moreover, the analysis of dendritic spine morphology did not show differences between mutant and control animals. DISCUSSION Despite the observed increase in newborn cells in the subventricular zone (SVZ) after riluzole administration, our study did not show any differences between riluzole-treated and non-treated mutants, revealing a similar extent of the neurodegenerative phenotype evaluated in 13-week-old TIF-IAD1RCre animals. This could be due to either the treatment paradigm (relatively low dose of riluzole used for this study) or the possibility that the effects were simply too weak to have any functional meaning. Nevertheless, this study is in line with others that question the effectiveness of riluzole in animal models and raise concerns about the utility of this drug due to its rather modest clinical efficacy.
Collapse
Affiliation(s)
- Grzegorz Kreiner
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Katarzyna Rafa-Zabłocka
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Piotr Chmielarz
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Monika Bagińska
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| | - Irena Nalepa
- Institute of Pharmacology, Polish Academy of Sciences, Dept. Brain Biochemistry, Kraków, Poland
| |
Collapse
|
17
|
Lu H, Le WD, Xie YY, Wang XP. Current Therapy of Drugs in Amyotrophic Lateral Sclerosis. Curr Neuropharmacol 2016; 14:314-21. [PMID: 26786249 PMCID: PMC4876587 DOI: 10.2174/1570159x14666160120152423] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/16/2015] [Accepted: 10/09/2015] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), commonly termed as motor neuron disease (MND) in UK, is a chronically lethal disorder among the neurodegenerative diseases, meanwhile. ALS is basically irreversible and progressive deterioration of upper and lower motor neurons in the motor cortex, brain stem and medulla spinalis. Riluzole, used for the treatment of ALS, was demonstrated to slightly delay the initiation of respiratory dysfunction and extend the median survival of patients by a few months. In this study, the key biochemical defects were discussed, such as: mutant Cu/Zn superoxide dismutase, mitochondrial protectants, and anti-excitotoxic/ anti-oxidative / anti-inflammatory/ anti-apoptotic agents, so the related drug candidates that have been studied in ALS models would possibly be further used in ALS patients.
Collapse
Affiliation(s)
| | | | | | - Xiao-Ping Wang
- Department of Neurology, Shanghai First People's Hospital , Shanghai Jiao-Tong University, China, 200080.
| |
Collapse
|
18
|
Abdoulaye IA, Guo YJ. A Review of Recent Advances in Neuroprotective Potential of 3-N-Butylphthalide and Its Derivatives. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5012341. [PMID: 28053983 PMCID: PMC5178327 DOI: 10.1155/2016/5012341] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/30/2016] [Accepted: 11/01/2016] [Indexed: 01/26/2023]
Abstract
The research of alternative treatment for ischemic stroke and degenerative diseases has always been a priority in neurology. 3-N-Butylphthalide (NBP), a family of compounds initially isolated from the seeds of Apium graveolens Linn., has shown significant neuroprotective effects. Previous extensive studies have demonstrated that NBP promotes a better poststroke outcome and exerts a multitargeted action on several mechanisms, from oxidative stress to mitochondrial dysfunction to apoptosis to inflammation. Additionally, recent findings on several neurological disorders have shown that NBP's beneficial effects extend beyond the management of stroke. However, despite the increasing number of studies toward a better understanding and the rapid advances made in therapeutic options, to date, dl-3-N-butylphthalide, a synthetic variation of l-3-N-butylphthalide, remains the only clinically approved anti-ischemic agent in China, stressing the difficulties for a viable and effective transition from experimental to clinical practice. Events indicate that NBP, due to its multitargeted effect and the adaptability of its basic structure, can be an important game changer and a precursor to a whole new therapeutic approach to several neurological conditions. The present review discusses recent advances pertaining to the neuroprotective mechanisms of NBP-derived compounds and the possibility of their clinical implementation in the management of various neurological conditions.
Collapse
Affiliation(s)
- Idriss Ali Abdoulaye
- Department of Neurology, The Southeast University Affiliated Zhong Da Hospital, No. 87 Dingjiaqiao, Nanjing, Jiangsu Province 210009, China
| | - Yi Jing Guo
- Department of Neurology, The Southeast University Affiliated Zhong Da Hospital, No. 87 Dingjiaqiao, Nanjing, Jiangsu Province 210009, China
| |
Collapse
|
19
|
Fay JM, Zhu C, Proctor EA, Tao Y, Cui W, Ke H, Dokholyan NV. A Phosphomimetic Mutation Stabilizes SOD1 and Rescues Cell Viability in the Context of an ALS-Associated Mutation. Structure 2016; 24:1898-1906. [PMID: 27667694 PMCID: PMC5093072 DOI: 10.1016/j.str.2016.08.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/29/2016] [Accepted: 08/10/2016] [Indexed: 01/01/2023]
Abstract
The majority of amyotrophic lateral sclerosis (ALS)-related mutations in the enzyme Cu,Zn superoxide dismutase (SOD1), as well as a post-translational modification, glutathionylation, destabilize the protein and lead to a misfolded oligomer that is toxic to motor neurons. The biophysical role of another physiological SOD1 modification, T2-phosphorylation, has remained a mystery. Here, we find that a phosphomimetic mutation, T2D, thermodynamically stabilizes SOD1 even in the context of a strongly SOD1-destabilizing mutation, A4V, one of the most prevalent and aggressive ALS-associated mutations in North America. This stabilization protects against formation of toxic SOD oligomers and positively impacts motor neuron survival in cellular assays. We solve the crystal structure of T2D-SOD1 and explain its stabilization effect using discrete molecular dynamics (DMD) simulations. These findings imply that T2-phosphorylation may be a plausible innate cellular protection response against SOD1-induced cytotoxicity, and stabilizing the SOD1 native conformation might offer us viable pharmaceutical strategies against currently incurable ALS.
Collapse
Affiliation(s)
- James M Fay
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Program in Molecular and Cellular Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cheng Zhu
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elizabeth A Proctor
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Program in Molecular and Cellular Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yazhong Tao
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Wenjun Cui
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hengming Ke
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Program in Molecular and Cellular Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
20
|
Sakanyan V, Hulin P, Alves de Sousa R, Silva VAO, Hambardzumyan A, Nedellec S, Tomasoni C, Logé C, Pineau C, Roussakis C, Fleury F, Artaud I. Activation of EGFR by small compounds through coupling the generation of hydrogen peroxide to stable dimerization of Cu/Zn SOD1. Sci Rep 2016; 6:21088. [PMID: 26883293 PMCID: PMC4756678 DOI: 10.1038/srep21088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/14/2016] [Indexed: 11/09/2022] Open
Abstract
Activation of cell signaling by reactive chemicals and pollutants is an important issue for human health. It has been shown that lipophilic nitro-benzoxadiazole (NBD) compounds rapidly move across the plasma membrane and enhance Epidermal Growth Factor Receptor (EGFR) tyrosine phosphorylation in cancer cells. Unlike ligand-dependent activation, the mechanism of this induction relies on the generation of hydrogen peroxide, which is involved in the activation of the catalytic site of the receptor and the inactivation of protein tyrosine phosphatase PTP-1B. Production of H2O2 during redox transformation of NBD compounds is associated with the transition of a monomeric form of Cu/Zn superoxide dismutase 1 (SOD1) to stable dimers. The highly stable and functionally active SOD1 dimer, in the absence of adequate activities in downstream reactions, promotes the disproportionate production and accumulation of intracellular hydrogen peroxide shortly after exposure to NBD compounds. The intrinsic fluorescence of small compounds was used to demonstrate their binding to SOD1. Our data indicate that H2O2 and concomitantly generated electrophilic intermediates behave as independent entities, but all contribute to the biological reactivity of NBD compounds. This study opens a promising path to identify new biomarkers of oxidative/electrophilic stress in the progression of cancer and other diseases.
Collapse
Affiliation(s)
- Vehary Sakanyan
- IICiMed EA-1155, Faculté de Pharmacie, Faculté des Sciences et des Techniques, Université de Nantes, 2 rue de la Houssinière, 44322 Nantes, France.,ProtNeteomix, 29 rue de Provence, 44700 Orvault, France
| | - Philippe Hulin
- Plate-forme MicroPICell SFR Santé F. Bonamy-FED 4203/Inserm UMS016/CNRS UMS3556, 44007 Nantes, France
| | - Rodolphe Alves de Sousa
- UMR 8601, CNRS, Université Paris Descartes, PRES Paris cité, 45 rue des Saints-Pères, 75270 Paris Cedex06, France
| | - Viviane A O Silva
- UFIP CNRS UMR 6286, Mechanism and Regulation of DNA Repair team, Faculté des Sciences et des Techniques, Université de Nantes, 2 rue de la Houssinière, 44322 Nantes, France
| | | | - Steven Nedellec
- Plate-forme MicroPICell SFR Santé F. Bonamy-FED 4203/Inserm UMS016/CNRS UMS3556, 44007 Nantes, France
| | - Christophe Tomasoni
- IICiMed EA-1155, Faculté de Pharmacie, Faculté des Sciences et des Techniques, Université de Nantes, 2 rue de la Houssinière, 44322 Nantes, France
| | - Cédric Logé
- IICiMed EA-1155, Faculté de Pharmacie, Faculté des Sciences et des Techniques, Université de Nantes, 2 rue de la Houssinière, 44322 Nantes, France
| | - Charles Pineau
- Protim, Inserm U1085-Irset, Campus de Beaulieu, 35042 Rennes, France
| | - Christos Roussakis
- IICiMed EA-1155, Faculté de Pharmacie, Faculté des Sciences et des Techniques, Université de Nantes, 2 rue de la Houssinière, 44322 Nantes, France
| | - Fabrice Fleury
- UFIP CNRS UMR 6286, Mechanism and Regulation of DNA Repair team, Faculté des Sciences et des Techniques, Université de Nantes, 2 rue de la Houssinière, 44322 Nantes, France
| | - Isabelle Artaud
- UMR 8601, CNRS, Université Paris Descartes, PRES Paris cité, 45 rue des Saints-Pères, 75270 Paris Cedex06, France
| |
Collapse
|
21
|
Liang H, Bi S, Liu Y, Tang YN, Liu C. Theoretical study on Au(i)-catalyzed [2 + 2 + 2] cycloadditions of ynamides with two discrete nitriles. Org Biomol Chem 2016; 14:2637-44. [DOI: 10.1039/c5ob02568k] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The Au-catalyzed [2 + 2 + 2] cycloadditions of ynamides with two discrete nitriles were theoretically studied with the aid of DFT calculations.
Collapse
Affiliation(s)
- Haosheng Liang
- College of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu 273165
- P. R. China
| | - Siwei Bi
- College of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu 273165
- P. R. China
| | - Yuxia Liu
- College of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu 273165
- P. R. China
| | - Ya-nan Tang
- College of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu 273165
- P. R. China
| | - Congcong Liu
- College of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu 273165
- P. R. China
| |
Collapse
|
22
|
Amyotrophic lateral sclerosis models derived from human embryonic stem cells with different superoxide dismutase 1 mutations exhibit differential drug responses. Stem Cell Res 2015; 15:459-468. [DOI: 10.1016/j.scr.2015.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/15/2015] [Indexed: 12/14/2022] Open
|
23
|
Zou H, Limpert AS, Zou J, Dembo A, Lee PS, Grant D, Ardecky R, Pinkerton AB, Magnuson GK, Goldman ME, Rong J, Teriete P, Sheffler DJ, Reed JC, Cosford NDP. Benzodiazepinone derivatives protect against endoplasmic reticulum stress-mediated cell death in human neuronal cell lines. ACS Chem Neurosci 2015; 6:464-75. [PMID: 25544056 PMCID: PMC4368043 DOI: 10.1021/cn500297v] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
![]()
Endoplasmic
reticulum (ER) stress causes neuronal dysfunction followed
by cell death and is recognized as a feature of many neurodegenerative
diseases. Using a phenotypic screen, we recently identified benzodiazepinone
derivatives that reduce ER stress-mediated apoptosis in a rat neuronal
progenitor cell line (CSM14.1). Herein we describe how structure–activity
relationship (SAR) studies around these screening hits led to compounds
that display robust cytoprotective activity against thapsigargin-induced
ER stress in SH-SY5Y and H4 human neuronal cell lines. We demonstrate
that the most potent of these derivatives, compound 4hh, inhibits the activation of p38 MAP kinase (p38) and c-Jun N-terminal
kinase (JNK), protein kinases that are downstream signal effectors
of the unfolded protein response (UPR). Compound 4hh specifically
protects against thapsigargin-induced cell death and displays no protection
against other insults known to induce cellular stress or activate
p38. However, compound 4hh provides moderate inhibition
of p38 activity stimulated by compounds that disrupt calcium homeostasis.
Our data indicate that probe compound 4hh is a valuable
small molecule tool that can be used to investigate the effects of
ER stress on human neurons. This approach may provide the basis for
the future development of therapeutics for the treatment of neurodegenerative
diseases.
Collapse
Affiliation(s)
- Haixia Zou
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Allison S. Limpert
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Jiwen Zou
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Anna Dembo
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Pooi-San Lee
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Daniel Grant
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Robert Ardecky
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Anthony B. Pinkerton
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Gavin K. Magnuson
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Mark E. Goldman
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Juan Rong
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Peter Teriete
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Douglas J. Sheffler
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - John C. Reed
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| | - Nicholas D. P. Cosford
- Cell Death and Survival Networks Research Program,
NCI-Designated Cancer Center, and ‡Conrad Prebys Center for Chemical
Genomics, Sanford-Burnham Medical Research Institute, 10901 North
Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
24
|
Faravelli I, Bucchia M, Rinchetti P, Nizzardo M, Simone C, Frattini E, Corti S. Motor neuron derivation from human embryonic and induced pluripotent stem cells: experimental approaches and clinical perspectives. Stem Cell Res Ther 2014; 5:87. [PMID: 25157556 PMCID: PMC4100331 DOI: 10.1186/scrt476] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Motor neurons are cells located in specific areas of the central nervous system, such as brain cortex (upper motor neurons), brain stem, and spinal cord (lower motor neurons), which maintain control over voluntary actions. Motor neurons are affected primarily by a wide spectrum of neurological disorders, generally indicated as motor neuron diseases (MNDs): these disorders share symptoms related to muscular atrophy and paralysis leading to death. No effective treatments are currently available. Stem cell-derived motor neurons represent a promising research tool in disease modeling, drug screening, and development of therapeutic approaches for MNDs and spinal cord injuries. Directed differentiation of human pluripotent stem cells - human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) - toward specific lineages is the first crucial step in order to extensively employ these cells in early human development investigation and potential clinical applications. Induced pluripotent stem cells (iPSCs) can be generated from patients' own somatic cells (for example, fibroblasts) by reprogramming them with specific factors. They can be considered embryonic stem cell-like cells, which express stem cell markers and have the ability to give rise to all three germ layers, bypassing the ethical concerns. Thus, hiPSCs constitute an appealing alternative source of motor neurons. These motor neurons might be a great research tool, creating a model for investigating the cellular and molecular interactions underlying early human brain development and pathologies during neurodegeneration. Patient-specific iPSCs may also provide the premises for autologous cell replacement therapies without related risks of immune rejection. Here, we review the most recent reported methods by which hESCs or iPSCs can be differentiated toward functional motor neurons with an overview on the potential clinical applications.
Collapse
|
25
|
Karad SN, Liu RS. Regiocontrolled Gold-Catalyzed [2+2+2] Cycloadditions of Ynamides with Two Discrete Nitriles to Construct 4-Aminopyrimidine Cores. Angew Chem Int Ed Engl 2014; 53:9072-6. [DOI: 10.1002/anie.201405312] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Indexed: 11/10/2022]
|
26
|
Karad SN, Liu RS. Regiocontrolled Gold-Catalyzed [2+2+2] Cycloadditions of Ynamides with Two Discrete Nitriles to Construct 4-Aminopyrimidine Cores. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201405312] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Comparison of Two Old Phytochemicals versus Two Newly Researched Plant-Derived Compounds: Potential for Brain and Other Relevant Ailments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:682717. [PMID: 24949079 PMCID: PMC4034649 DOI: 10.1155/2014/682717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/31/2014] [Accepted: 04/15/2014] [Indexed: 11/28/2022]
Abstract
Among hundreds of formulae of Chinese herbal prescriptions and recently extracted active components from the herbs, some of which had demonstrated their functions on nervous system. For the last decade or more, Gingko biloba and Polygala tenuifolia were widely studied for their beneficial effects against damage to the brain. Two compounds extracted from Apium graveolens and Rhizoma coptidis, butylphthalide and berberine, respectively, received much attention recently as potential neuroprotective agents. In this review, the two traditionally used herbs and the two relatively new compounds will be discussed with regard to their potential advantages in alleviating brain and other relevant ailments.
Collapse
|
28
|
Limpert AS, Cosford NDP. Translational enhancers of EAAT2: therapeutic implications for neurodegenerative disease. J Clin Invest 2014; 124:964-7. [PMID: 24569369 DOI: 10.1172/jci74608] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glutamate excitotoxicity contributes to the neuronal injury and death associated with many neurodegenerative diseases. The glutamate transporter EAAT2, which is primarily localized on astrocytic processes, facilitates glutamate clearance from synapses, thus preventing neuronal damage. In this issue of the JCI, Kong et al. characterize a compound that upregulates EAAT2 translation, thereby increasing glutamate uptake by glial cells. Furthermore, this strategy for alleviating excitotoxicity was found to be beneficial in mouse models of both amyotrophic lateral sclerosis (ALS) and epilepsy, suggesting that future development in this chemical series may lead to much-needed treatments for these disorders.
Collapse
|
29
|
Tanaka K, Kanno T, Yanagisawa Y, Yasutake K, Inoue S, Hirayama N, Ikeda JE. A novel acylaminoimidazole derivative, WN1316, alleviates disease progression via suppression of glial inflammation in ALS mouse model. PLoS One 2014; 9:e87728. [PMID: 24498180 PMCID: PMC3909264 DOI: 10.1371/journal.pone.0087728] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/27/2013] [Indexed: 12/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron degenerative disease. Given that oxidative stress and resulting chronic neuronal inflammation are thought to be central pathogenic, anti-oxidative agents and modulators of neuronal inflammation could be potential therapies for ALS. We report here that the novel small molecular compound, 2-[mesityl(methyl)amino]-N-[4-(pyridin-2-yl)-1H-imidazol-2-yl] acetamide trihydrochloride (WN1316) selectively suppresses oxidative stress-induced cell death and neuronal inflammation in the late-stage ALS mice. WN1316 has high blood-brain-barrier permeability and water solubility, and boosts both neuronal apoptosis inhibitory protein (NAIP) and NF-E2-related factor 2 (Nrf2) which governed glutathione (GSH)-related anti-oxidation pathway protecting motor neurons against oxidative injuries. Post-onset oral administration of low dose (1-100 µg/kg/day) WN1316 in ALS(SOD1(H46R)) and ALS(SOD1(G93A)) mice resulted in sustained improved motor function and post onset survival rate. Immunohistochemical analysis revealed less DNA oxidative damage and motor neuronal inflammation as well as repression of both microgliosis and astrocytosis, concomitant down regulation of interleukin-1β and inducible nitric oxide synthase, and preservation of the motoneurons in anterior horn of lumbar spinal cord and skeletal muscle (quadriceps femoris). Thus, WN1316 would be a novel therapeutic agent for ALS.
Collapse
Affiliation(s)
- Kazunori Tanaka
- NGP Biomedical Research Institute, Neugen Pharma Inc., Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Takuya Kanno
- NGP Biomedical Research Institute, Neugen Pharma Inc., Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Yoshiko Yanagisawa
- NGP Biomedical Research Institute, Neugen Pharma Inc., Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Kaori Yasutake
- NGP Biomedical Research Institute, Neugen Pharma Inc., Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Satoshi Inoue
- Wakunaga Pharmaceutical Co. Ltd., Akitakada, Hiroshima, Japan
| | - Noriaki Hirayama
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Joh-E Ikeda
- NGP Biomedical Research Institute, Neugen Pharma Inc., Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Department of Molecular Neurology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- Apoptosis Research Centre, Children’s Hospital of Eastern Ontario, Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
30
|
Cristóvão JS, Leal SS, Cardoso I, Gomes CM. Small molecules present in the cerebrospinal fluid metabolome influence superoxide dismutase 1 aggregation. Int J Mol Sci 2013; 14:19128-45. [PMID: 24048249 PMCID: PMC3794824 DOI: 10.3390/ijms140919128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 08/28/2013] [Accepted: 08/30/2013] [Indexed: 12/13/2022] Open
Abstract
Superoxide dismutase 1 (SOD1) aggregation is one of the pathological markers of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. The underlying molecular grounds of SOD1 pathologic aggregation remains obscure as mutations alone are not exclusively the cause for the formation of protein inclusions. Thus, other components in the cell environment likely play a key role in triggering SOD1 toxic aggregation in ALS. Recently, it was found that ALS patients present a specific altered metabolomic profile in the cerebrospinal fluid (CSF) where SOD1 is also present and potentially interacts with metabolites. Here we have investigated how some of these small molecules affect apoSOD1 structure and aggregation propensity. Our results show that as co-solvents, the tested small molecules do not affect apoSOD1 thermal stability but do influence its tertiary interactions and dynamics, as evidenced by combined biophysical analysis and proteolytic susceptibility. Moreover, these compounds influence apoSOD1 aggregation, decreasing nucleation time and promoting the formation of larger and less soluble aggregates, and in some cases polymeric assemblies apparently composed by spherical species resembling the soluble native protein. We conclude that some components of the ALS metabolome that shape the chemical environment in the CSF may influence apoSOD1 conformers and aggregation.
Collapse
Affiliation(s)
- Joana S. Cristóvão
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, EAN, Oeiras 2784-505, Portugal; E-Mails: (J.S.C.); (S.S.L.)
| | - Sónia S. Leal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, EAN, Oeiras 2784-505, Portugal; E-Mails: (J.S.C.); (S.S.L.)
| | - Isabel Cardoso
- Molecular Neurobiology Unit, Instituto de Biologia Molecular e Celular, Rua do Campo Alegre, 823, Porto 4150-180, Portugal; E-Mail:
- Escola Superior de Tecnologia da Saúde do Porto, Instituto Politécnico do Porto, Rua Valente Perfeito, 322, Vila Nova de Gaia 4400-330, Portugal
| | - Cláudio M. Gomes
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, EAN, Oeiras 2784-505, Portugal; E-Mails: (J.S.C.); (S.S.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +351-21-446-9332; Fax: +351-21-441-1277
| |
Collapse
|