1
|
López-García P, Tejero-Ojeda MM, Vaquero ME, Carrión-Vázquez M. Current amyloid inhibitors: Therapeutic applications and nanomaterial-based innovations. Prog Neurobiol 2025; 247:102734. [PMID: 40024279 DOI: 10.1016/j.pneurobio.2025.102734] [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: 09/03/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Amyloid proteins have long been in the spotlight for being involved in many degenerative diseases including Alzheimer´s, Parkinson´s or type 2 diabetes, which currently cannot be prevented and for which there is no effective treatment or cure. Here we provide a comprehensive review of inhibitors that act directly on the amyloidogenic pathway (at the monomer, oligomer or fibril level) of key pathological amyloids, focusing on the most representative amyloid-related diseases. We discuss the latest advances in preclinical and clinical trials, focusing on cutting-edge developments, particularly on nanomaterials-based inhibitors, which offer unprecedented opportunities to address the complexity of protein misfolding disorders and are revolutionizing the landscape of anti-amyloid therapeutics. Notably, nanomaterials are impacting critical areas such as bioavailability, penetrability and functionality of compounds currently used in biomedicine, paving the way for more specific therapeutic solutions tailored to various amyloid-related diseases. Finally, we highlight the window of opportunity opened by comparative analysis with so-called functional amyloids for the development of innovative therapeutic approaches for these devastating diseases.
Collapse
|
2
|
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
|
3
|
Rani A, Saini V, Patra P, Prashar T, Pandey RK, Mishra A, Jha HC. Epigallocatechin Gallate: A Multifaceted Molecule for Neurological Disorders and Neurotropic Viral Infections. ACS Chem Neurosci 2023; 14:2968-2980. [PMID: 37590965 DOI: 10.1021/acschemneuro.3c00368] [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: 08/19/2023] Open
Abstract
Epigallocatechin-3-gallate (EGCG), a polyphenolic moiety found in green tea extracts, exhibits pleiotropic bioactivities to combat many diseases including neurological ailments. These neurological diseases include Alzheimer's disease, multiple sclerosis, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. For instance, in the case of Alzheimer's disease, the formation of a β-sheet in the region of the 10th-21st amino acids was significantly reduced in EGCG-induced oligomeric samples of Aβ40. Its interference induces the formation of Aβ structures with an increase in intercenter-of-mass distances, reduction in interchain/intrachain contacts, reduction in β-sheet propensity, and increase in α-helix. Besides, numerous neurotropic viruses are known to instigate or aggravate neurological ailments. It exerts an effect on the oxidative damage caused in neurodegenerative disorders by acting on GSK3-β, PI3K/Akt, and downstream signaling pathways via caspase-3 and cytochrome-c. EGCG also diminishes these viral-mediated effects, such as EGCG delayed HSV-1 infection by blocking the entry for virions, inhibitory effects on NS3/4A protease or NS5B polymerase of HCV and potent inhibitor of ZIKV NS2B-NS3pro/NS3 serine protease (NS3-SP). It showed a reduction in the neurotoxic properties of HIV-gp120 and Tat in the presence of IFN-γ. EGCG also involves numerous viral-mediated inflammatory cascades, such as JAK/STAT. Nonetheless, it also inhibits the Epstein-Barr virus replication protein (Zta and Rta). Moreover, it also impedes certain viruses (influenza A and B strains) by hijacking the endosomal and lysosomal compartments. Therefore, the current article aims to describe the importance of EGCG in numerous neurological diseases and its inhibitory effect against neurotropic viruses.
Collapse
Affiliation(s)
- Annu Rani
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| | - Vaishali Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| | - Priyanka Patra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| | - Tanish Prashar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Karwar, 342030, Jodhpur India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| |
Collapse
|
4
|
Sadasivan J, Hyrina A, DaSilva R, Jan E. An Insect Viral Protein Disrupts Stress Granule Formation in Mammalian Cells. J Mol Biol 2023; 435:168042. [PMID: 36898623 DOI: 10.1016/j.jmb.2023.168042] [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: 08/29/2022] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023]
Abstract
Stress granules (SGs) are cytosolic RNA-protein aggregates assembled during stress-induced translation arrest. Virus infection, in general, modulates and blocks SG formation. We previously showed that the model dicistrovirus Cricket paralysis virus (CrPV) 1A protein blocks stress granule formation in insect cells, which is dependent on a specific arginine 146 residue. CrPV-1A also inhibits SG formation in mammalian cells suggesting that this insect viral protein may be acting on a fundamental process that regulates SG formation. The mechanism underlying this process is not fully understood. Here, we show that overexpression of wild-type CrPV-1A, but not the CrPV-1A(R146A) mutant protein, inhibits distinct SG assembly pathways in HeLa cells. CrPV-1A mediated SG inhibition is independent of the Argonaute-2 (Ago-2) binding domain and the E3 ubiquitin ligase recruitment domain. CrPV-1A expression leads to nuclear poly(A)+ RNA accumulation and is correlated with the localization of CrPV-1A to the nuclear periphery. Finally, we show that the overexpression of CrPV-1A blocks FUS and TDP-43 granules, which are pathological hallmarks of neurodegenerative diseases. We propose a model whereby CrPV-1A expression in mammalian cells blocks SG formation by depleting cytoplasmic mRNA scaffolds via mRNA export inhibition. CrPV-1A provides a new molecular tool to study RNA-protein aggregates and potentially uncouple SG functions.
Collapse
Affiliation(s)
- Jibin Sadasivan
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada. https://twitter.com/@jibin_sadasivan
| | - Anastasia Hyrina
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rachel DaSilva
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eric Jan
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
5
|
The current state of amyloidosis therapeutics and the potential role of fluorine in their treatment. Biochimie 2022; 202:123-135. [PMID: 35963462 DOI: 10.1016/j.biochi.2022.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/22/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022]
Abstract
Amyloidosis, commonly known as amyloid-associated diseases, is characterized by improperly folded proteins accumulating in tissues and eventually causing organ damage, which is linked to several disorders ranging from neurodegenerative to peripheral diseases. It has an enormous societal and financial impact on the global health sector. Due to the complexity of protein misfolding and intertwined aggregation, there are no effective disease-modifying medications at present, and the condition is likely mis/non-diagnosed half of the time. Nonetheless, over the last two decades, substantial research into aggregation processes has revealed the possibilities of new intervention approaches. On the other hand, fluorine has been a rising star in therapeutic development for numerous neurodegenerative illnesses and other peripheral diseases. In this study, we revised and emphasized the possible significance of fluorine-modified therapeutic molecules and fluorine-modified nanoparticles (NPs) in the modulation of amyloidogenic proteins, including insulin, amyloid beta peptide (Aβ), prion protein (PrP), transthyretin (TTR) and Huntingtin (htt).
Collapse
|
6
|
Kolkwitz PE, Mohrlüder J, Willbold D. Inhibition of Polyglutamine Misfolding with D-Enantiomeric Peptides Identified by Mirror Image Phage Display Selection. Biomolecules 2022; 12:biom12020157. [PMID: 35204656 PMCID: PMC8961585 DOI: 10.3390/biom12020157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Nine heritable diseases are known that are caused by unphysiologically elongated polyglutamine tracts in human proteins leading to misfolding, aggregation and neurodegeneration. Current therapeutic strategies include efforts to inhibit the expression of the respective gene coding for the polyglutamine-containing proteins. There are, however, concerns that this may interfere with the physiological function of the respective protein. We aim to stabilize the protein’s native conformation by D-enantiomeric peptide ligands to prevent misfolding and aggregation, shift the equilibrium between aggregates and monomers towards monomers and dissolve already existing aggregates into non-toxic and functional monomers. Here, we performed a mirror image phage display selection on the polyglutamine containing a fragment of the androgen receptor. An elongated polyglutamine tract in the androgen receptor causes spinal and bulbar muscular atrophy (SBMA). The selected D-enantiomeric peptides were tested for their ability to inhibit polyglutamine-induced androgen receptor aggregation. We identified D-enantiomeric peptide QF2D-2 (sqsqwstpqGkwshwprrr) as the most promising candidate. It binds to an androgen receptor fragment with 46 consecutive glutamine residues and decelerates its aggregation, even in seeded experiments. Therefore, QF2D-2 may be a promising drug candidate for SBMA treatment or even for all nine heritable polyglutamine diseases, since its aggregation-inhibiting property was shown also for a more general polyglutamine target.
Collapse
Affiliation(s)
- Pauline Elisabeth Kolkwitz
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (P.E.K.); (J.M.)
| | - Jeannine Mohrlüder
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (P.E.K.); (J.M.)
| | - Dieter Willbold
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (P.E.K.); (J.M.)
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence:
| |
Collapse
|
7
|
Wahyuningtyas D, Chen WH, He RY, Huang YA, Tsao CK, He YJ, Yu CY, Lu PC, Chen YC, Wang SH, Ng KC, Po-Wen Chen B, Wei PK, Shie JJ, Kuo CH, Sun YH, Jen-Tse Huang J. Polyglutamine-Specific Gold Nanoparticle Complex Alleviates Mutant Huntingtin-Induced Toxicity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:60894-60906. [PMID: 34914364 DOI: 10.1021/acsami.1c18754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Huntington's disease (HD) belongs to protein misfolding disorders associated with polyglutamine (polyQ)-rich mutant huntingtin (mHtt) protein inclusions. Currently, it is indicated that the aggregation of polyQ-rich mHtt participates in neuronal toxicity and dysfunction. Here, we designed and synthesized a polyglutamine-specific gold nanoparticle (AuNP) complex, which specifically targeted mHtt and alleviated its toxicity. The polyglutamine-specific AuNPs were prepared by decorating the surface of AuNPs with an amphiphilic peptide (JLD1) consisting of both polyglutamine-binding sequences and negatively charged sequences. By applying the polyQ aggregation model system, we demonstrated that AuNPs-JLD1 dissociated the fibrillary aggregates from the polyQ peptide and reduced its β-sheet content in a concentration-dependent manner. By further integrating polyethyleneimine (PEI) onto AuNPs-JLD1, we generated a complex (AuNPs-JLD1-PEI). We showed that this complex could penetrate cells, bind to cytosolic mHtt proteins, dissociate mHtt inclusions, reduce mHtt oligomers, and ameliorate mHtt-induced toxicity. AuNPs-JLD1-PEI was also able to be transported to the brain and improved the functional deterioration in the HD Drosophila larva model. Our results revealed the feasibility of combining AuNPs, JLD1s, and cell-penetrating polymers against mHtt protein aggregation and oligomerization, which hinted on the early therapeutic strategies against HD.
Collapse
Affiliation(s)
- Devi Wahyuningtyas
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Science Building 2, 1001 Ta Hsueh Road, Hsinchu 300, Taiwan
| | - Wen-Hao Chen
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Ruei-Yu He
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Yung-An Huang
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Chia-Kang Tsao
- Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Yu-Jung He
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Chu-Yi Yu
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Po-Chao Lu
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
- Department and Graduate Institute of Pharmacology, National Taiwan University, 11F, No. 1, Section 1, Ren'ai Road, Zhongzheng District, Taipei 10051, Taiwan
| | - Yu-Cai Chen
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Sheng-Hann Wang
- Research Center for Applied Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 115, Taiwan
| | - Ka Chon Ng
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Bryan Po-Wen Chen
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 115, Taiwan
| | - Jiun-Jie Shie
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Chun-Hong Kuo
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Y Henry Sun
- Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
| | - Joseph Jen-Tse Huang
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang District, Taipei 11529, Taiwan
- Department of Applied Chemistry, National Chiayi University, No. 300, University Road, Chiayi 600, Taiwan
- Neuroscience Program of Academia Sinica, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei 11529, Taiwan
| |
Collapse
|
8
|
Belwal VK, Vijayakumar A, Chaudhary N. Inhibition of huntingtin aggregation by its N-terminal 17-residue peptide and its analogs. Arch Biochem Biophys 2021; 712:109033. [PMID: 34534539 DOI: 10.1016/j.abb.2021.109033] [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: 04/01/2021] [Revised: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
The N-terminal 17-residue stretch of huntingtin (httN17) folds into an amphipathic α-helix. The httN17-harboring polyQ peptides form oligomers that are mediated via the assembly of the httN17 α-helices. The oligomerization results in higher local concentration of the polyglutamine (polyQ) region, thereby facilitating amyloid formation. The httN17 co-assembles with the httN17-harbouring polyQ peptides, thereby reducing the local polyQ concentration, and consequently inhibiting aggregation. This study presents the aggregation inhibition of the exon I region of pathogenic huntingtin by httN17 and its analogs. The C-terminal amidation of httN17 is found to be essential for activity. The httN17 peptides with free amino terminus and the acetylated amino terminus possess comparable activity. The httN17 analog, wherein the Leu7 and Ala10 are substituted with 2-aminoisobutyric acid residues, exhibits significantly higher activity than the native httN17.
Collapse
Affiliation(s)
- Vinay Kumar Belwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781 039, India
| | - Aishwarya Vijayakumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781 039, India
| | - Nitin Chaudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781 039, India.
| |
Collapse
|
9
|
Li J, Gao G, Tang X, Yu M, He M, Sun T. Isomeric Effect of Nano-Inhibitors on Aβ 40 Fibrillation at The Nano-Bio Interface. ACS APPLIED MATERIALS & INTERFACES 2021; 13:4894-4904. [PMID: 33486955 DOI: 10.1021/acsami.0c21906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chemical and physical properties of nanobio interface substantially affect the conformational transitions of adjacent biomolecules. Previous studies have reported the chiral effect and charge effect of nanobio interface on the misfolding, aggregation, and fibrillation of amyloid protein. However, the isomeric effect of nanobio interface on protein/peptides amyloidosis is still unclear. Here, three isomeric nanobio interfaces were designed and fabricated based on the same sized gold nanoclusters (AuNCs) modified with 4-mercaptobenzoic acid (p-MBA), 3-mercaptobenzoic acid (m-MBA), and 2-mercaptobenzoic acid (o-MBA). Then three isomeric AuNCs were employed as models to explore the isomeric effect on the misfolding, aggregation, and fibrillation of Aβ40 at nanobio interfaces. Site-specific replacement experiments on the basis of theoretical analysis revealed the possible mechanism of Aβ40 interacting with isomeric ligands of AuNCs at the nanobio interfaces. The distance and orientation of -COOH group from the surface of AuNCs can affect the electrostatic interaction between isomeric ligands and the positively charged residues (R5, K16, and K28) of Aβ40, which may affect the inhibition efficiency of isomeric AuNCs on protein amyloidosis. Actually, the amyloid fibrillation kinetics results together with atomic force microscope (AFM) images, dynamic light scattering (DLS) results and circular dichroism (CD) spectra indeed proved that all the three isomeric AuNCs could inhibit the misfolding, aggregation and fibrillation of Aβ40 in a dose-dependent manner, and the inhibition efficiency was definitely different from each other. The inhibition efficiency of o-MBA-AuNCs was higher than that of m-MBA-AuNCs and p-MBA-AuNCs at the same dosage. These results provide an insight for isomeric effect at nanobio interfaces, and open an avenue for structure-based nanodrug design target Alzheimer's disease (AD) and even other protein conformational diseases.
Collapse
Affiliation(s)
- Jianhang Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, No.122 Luoshi Road, Wuhan 430070, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, No.122 Luoshi Road, Wuhan 430070, China
| | - Xintong Tang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, No.122 Luoshi Road, Wuhan 430070, China
| | - Meng Yu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, No.122 Luoshi Road, Wuhan 430070, China
| | - Meng He
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, No.122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, No.122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
10
|
Niewiadomska-Cimicka A, Trottier Y. Molecular Targets and Therapeutic Strategies in Spinocerebellar Ataxia Type 7. Neurotherapeutics 2019; 16:1074-1096. [PMID: 31432449 PMCID: PMC6985300 DOI: 10.1007/s13311-019-00778-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a rare autosomal dominant neurodegenerative disorder characterized by progressive neuronal loss in the cerebellum, brainstem, and retina, leading to cerebellar ataxia and blindness as major symptoms. SCA7 is due to the expansion of a CAG triplet repeat that is translated into a polyglutamine tract in ATXN7. Larger SCA7 expansions are associated with earlier onset of symptoms and more severe and rapid disease progression. Here, we summarize the pathological and genetic aspects of SCA7, compile the current knowledge about ATXN7 functions, and then focus on recent advances in understanding the pathogenesis and in developing biomarkers and therapeutic strategies. ATXN7 is a bona fide subunit of the multiprotein SAGA complex, a transcriptional coactivator harboring chromatin remodeling activities, and plays a role in the differentiation of photoreceptors and Purkinje neurons, two highly vulnerable neuronal cell types in SCA7. Polyglutamine expansion in ATXN7 causes its misfolding and intranuclear accumulation, leading to changes in interactions with native partners and/or partners sequestration in insoluble nuclear inclusions. Studies of cellular and animal models of SCA7 have been crucial to unveil pathomechanistic aspects of the disease, including gene deregulation, mitochondrial and metabolic dysfunctions, cell and non-cell autonomous protein toxicity, loss of neuronal identity, and cell death mechanisms. However, a better understanding of the principal molecular mechanisms by which mutant ATXN7 elicits neurotoxicity, and how interconnected pathogenic cascades lead to neurodegeneration is needed for the development of effective therapies. At present, therapeutic strategies using nucleic acid-based molecules to silence mutant ATXN7 gene expression are under development for SCA7.
Collapse
Affiliation(s)
- Anna Niewiadomska-Cimicka
- Institute of Genetic and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR7104), Institut National de la Santé et de la Recherche Médicale (U1258), University of Strasbourg, Illkirch, France
| | - Yvon Trottier
- Institute of Genetic and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR7104), Institut National de la Santé et de la Recherche Médicale (U1258), University of Strasbourg, Illkirch, France.
| |
Collapse
|
11
|
Wahyuningtyas D, Chen WH, Huang CH, He YJ, Huang JJT. Biocompatible Inhibitor Based on Chitosan and Amphiphilic Peptide against Mutant Huntingtin Toxicity. Chembiochem 2019; 20:2133-2140. [PMID: 31166067 DOI: 10.1002/cbic.201900242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Indexed: 12/15/2022]
Abstract
Huntington's disease (HD) is classified as a protein-misfolding disease correlated with the mutant Huntingtin (mHtt) protein with abnormally expanded polyglutamine (polyQ) domains. Because no effective drugs have yet been reported, attempts to develop better therapy to delay the age of onset are in urgent demand. In this study, an amphiphilic peptide consisting of negatively charged hexaglutamic acid and a stretch of decaglutamine (E6 Q10 ) was chemically synthesized as an inhibitor against polyQ and mHtt toxicity. It is found that E6 Q10 selfassembles into spherical vesicles, as shown by means of TEM, cryoelectron microscopy, and dynamic light scattering. Assembled E6 Q10 prevented the polyQ-rich peptide (KKWQ20 AKK) from forming amyloid fibrils. To enable the cell-penetration ability of E6 Q10 , the E6 Q10 ⋅chitosan complex was generated. It is demonstrated that the complex penetrates cells, interferes with the mHtt oligomerization and aggregation process, and prevents mHtt cytotoxicity. By combining positively charged chitosan and amphiphilic peptides with a negatively charge moiety, a new strategy is provided to develop biocompatible and biodegradable inhibitors against mHtt toxicity.
Collapse
Affiliation(s)
- Devi Wahyuningtyas
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 115, Taiwan.,Sustainable Chemical Science and Technology Program, Taiwan International Graduate Program, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 115, Taiwan.,Department of Applied Chemistry, National Chiao Tung University, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300, Taiwan
| | - Wen-Hao Chen
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 115, Taiwan
| | - Chen-Han Huang
- Department of Biomedical Sciences and Engineering, National Central University, No. 300, Zhongda Road, Zhongli, Taoyuan, 32001, Taiwan
| | - Yu-Jung He
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 115, Taiwan
| | - Joseph Jen-Tse Huang
- Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 115, Taiwan
| |
Collapse
|
12
|
Yang M, Zhang Q, Wang Q, Sørensen KK, Boesen JT, Ma SY, Jensen KJ, Kwan KM, Ngo JCK, Chan HYE, Zuo Z. Brain-Targeting Delivery of Two Peptidylic Inhibitors for Their Combination Therapy in Transgenic Polyglutamine Disease Mice via Intranasal Administration. Mol Pharm 2018; 15:5781-5792. [PMID: 30392378 DOI: 10.1021/acs.molpharmaceut.8b00938] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Polyglutamine diseases are a set of progressive neurodegenerative disorders caused by misfolding and aggregation of mutant CAG RNA and polyglutamin protein. To date, there is a lack of effective therapeutics that can counteract the polyglutamine neurotoxicity. Two peptidylic inhibitors, QBP1 and P3, targeting the protein and RNA toxicities, respectively, have been previously demonstrated by us with combinational therapeutic effects on the Drosophila polyglutamine disease model. However, their therapeutic efficacy has never been investigated in vivo in mammals. The current study aims to (a) develop a brain-targeting delivery system for both QBP1 and L1P3V8 (a lipidated variant of P3 with improved stability) and (b) evaluate their therapeutic effects on the R6/2 transgenic mouse model of polyglutamine disease. Compared with intravenous administration, intranasal administration of QBP1 significantly increased its brain-to-plasma ratio. In addition, employment of a chitosan-containing in situ gel for the intranasal administration of QBP1 notably improved its brain concentration for up to 10-fold. Further study on intranasal cotreatment with the optimized formulation of QBP1 and L1P3V8 in mice found no interference on the brain uptake of each other. Subsequent efficacy evaluation of 4-week daily QBP1 (16 μmol/kg) and L1P3V8 (6 μmol/kg) intranasal cotreatment in the R6/2 mice demonstrated a significant improvement on the motor coordination and explorative behavior of the disease mice, together with a full suppression on the RNA- and protein-toxicity markers in their brains. In summary, the current study developed an efficient intranasal cotreatment of the two peptidylic inhibitors, QBP1 and L1P3V8, for their brain-targeting, and such a novel therapeutic strategy was found to be effective on a transgenic polyglutamine disease mouse model.
Collapse
Affiliation(s)
- Mengbi Yang
- School of Pharmacy , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Qian Zhang
- School of Life Sciences , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Qianwen Wang
- School of Pharmacy , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Kasper K Sørensen
- Department of Chemistry , University of Copenhagen , Thorvaldsensvej 40 , 1871 Frederiksberg , Denmark
| | - Josephine T Boesen
- Department of Chemistry , University of Copenhagen , Thorvaldsensvej 40 , 1871 Frederiksberg , Denmark
| | - Sum Yi Ma
- School of Life Sciences , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Knud J Jensen
- Department of Chemistry , University of Copenhagen , Thorvaldsensvej 40 , 1871 Frederiksberg , Denmark
| | - Kin Ming Kwan
- School of Life Sciences , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China.,Partner State Key Laboratory of Agrobiotechnology , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Jacky Chi Ki Ngo
- School of Life Sciences , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Ho Yin Edwin Chan
- School of Life Sciences , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China.,Gerald Choa Neuroscience Centre , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| | - Zhong Zuo
- School of Pharmacy , The Chinese University of Hong Kong , Shatin, Hong Kong , SAR , China
| |
Collapse
|
13
|
Takeuchi T. Non-cell Autonomous Maintenance of Proteostasis by Molecular Chaperones and Its Molecular Mechanism. Biol Pharm Bull 2018; 41:843-849. [PMID: 29863073 DOI: 10.1248/bpb.b18-00141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Molecular chaperones have essential roles in cell survival, to prevent misfolding, aggregation, and aberrant accumulation of cellular proteins, and thus to maintain protein homeostasis (proteostasis). However, recent studies using animal models suggest that transcriptional upregulation of molecular chaperones in response to various types of stresses does not ubiquitously occur in all cells and tissues, but is a cell type-specific event. The imbalanced response to stresses between cells and tissues has been pointed out since more than 30 years ago, but the molecular basis as to how organisms maintain proteostasis in all cells, especially cells deficient for chaperone induction, remains unknown. In this review, I introduce the non-cell autonomous function of molecular chaperones that has been suggested in animal studies, especially focusing on our recent findings, and discuss the possibility that the non-cell autonomous function might provide a potential explanation as to how organisms would maintain proteostasis despite the imbalanced stress response between cells and tissues. Further elucidation of the molecular basis underlying the non-cell autonomous function of molecular chaperones would provide not only better understanding as to how organisms maintain proteostasis but also important insights into the potential development of therapies and diagnostics for the currently intractable neurodegenerative diseases that are associated with protein misfolding and aggregation.
Collapse
Affiliation(s)
- Toshihide Takeuchi
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine.,JST, PRESTO
| |
Collapse
|
14
|
Protein aggregation: From background to inhibition strategies. Int J Biol Macromol 2017; 103:208-219. [DOI: 10.1016/j.ijbiomac.2017.05.048] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 01/01/2023]
|
15
|
Polyglutamine expansion diseases: More than simple repeats. J Struct Biol 2017; 201:139-154. [PMID: 28928079 DOI: 10.1016/j.jsb.2017.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/24/2017] [Accepted: 09/15/2017] [Indexed: 12/27/2022]
Abstract
Polyglutamine (polyQ) repeat-containing proteins are widespread in the human proteome but only nine of them are associated with highly incapacitating neurodegenerative disorders. The genetic expansion of the polyQ tract in disease-related proteins triggers a series of events resulting in neurodegeneration. The polyQ tract plays the leading role in the aggregation mechanism, but other elements modulate the aggregation propensity in the context of the full-length proteins, as implied by variations in the length of the polyQ tract required to trigger the onset of a given polyQ disease. Intrinsic features such as the presence of aggregation-prone regions (APRs) outside the polyQ segments and polyQ-flanking sequences, which synergistically participate in the aggregation process, are emerging for several disease-related proteins. The inherent polymorphic structure of polyQ stretches places the polyQ proteins in a central position in protein-protein interaction networks, where interacting partners may additionally shield APRs or reshape the aggregation course. Expansion of the polyQ tract perturbs the cellular homeostasis and contributes to neuronal failure by modulating protein-protein interactions and enhancing toxic oligomerization. Post-translational modifications further regulate self-assembly either by directly altering the intrinsic aggregation propensity of polyQ proteins, by modulating their interaction with different macromolecules or by modifying their withdrawal by the cell quality control machinery. Here we review the recent data on the multifaceted aggregation pathways of disease-related polyQ proteins, focusing on ataxin-3, the protein mutated in Machado-Joseph disease. Further mechanistic understanding of this network of events is crucial for the development of effective therapies for polyQ diseases.
Collapse
|
16
|
Escalona-Rayo O, Fuentes-Vázquez P, Leyva-Gómez G, Cisneros B, Villalobos R, Magaña JJ, Quintanar-Guerrero D. Nanoparticulate strategies for the treatment of polyglutamine diseases by halting the protein aggregation process. Drug Dev Ind Pharm 2017; 43:871-888. [PMID: 28142290 DOI: 10.1080/03639045.2017.1281949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Polyglutamine (polyQ) diseases are a class of neurodegenerative disorders that cause cellular dysfunction and, eventually, neuronal death in specific regions of the brain. Neurodegeneration is linked to the misfolding and aggregation of expanded polyQ-containing proteins, and their inhibition is one of major therapeutic strategies used commonly. However, successful treatment has been limited to date because of the intrinsic properties of therapeutic agents (poor water solubility, low bioavailability, poor pharmacokinetic properties), and difficulty in crossing physiological barriers, including the blood-brain barrier (BBB). In order to solve these problems, nanoparticulate systems with dimensions of 1-1000 nm able to incorporate small and macromolecules with therapeutic value, to protect and deliver them directly to the brain, have recently been developed, but their use for targeting polyQ disease-mediated protein misfolding and aggregation remains scarce. This review provides an update of the polyQ protein aggregation process and the development of therapeutic strategies for halting it. The main features that a nanoparticulate system should possess in order to enhance brain delivery are discussed, as well as the different types of materials utilized to produce them. The final part of this review focuses on the potential application of nanoparticulate system strategies to improve the specific and efficient delivery of therapeutic agents to the brain for the treatment of polyQ diseases.
Collapse
Affiliation(s)
- Oscar Escalona-Rayo
- a Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| | - Paulina Fuentes-Vázquez
- a Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| | - Gerardo Leyva-Gómez
- b Laboratory of Connective Tissue , CENIAQ, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra , Mexico City , Mexico
| | - Bulmaro Cisneros
- c Department of Genetics and Molecular Biology , CINVESTAV-IPN , Mexico City , Mexico
| | - Rafael Villalobos
- d División de Estudios de Posgrado (Tecnología Farmacéutica), Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| | - Jonathan J Magaña
- e Laboratory of Genomic Medicine, Department of Genetics , Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra , Mexico City , Mexico
| | - David Quintanar-Guerrero
- a Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| |
Collapse
|
17
|
Zhang Q, Tsoi H, Peng S, Li PP, Lau KF, Rudnicki DD, Ngo JCK, Chan HYE. Assessing a peptidylic inhibitor-based therapeutic approach that simultaneously suppresses polyglutamine RNA- and protein-mediated toxicities in patient cells and Drosophila. Dis Model Mech 2016; 9:321-34. [PMID: 26839389 PMCID: PMC4833327 DOI: 10.1242/dmm.022350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/27/2016] [Indexed: 02/03/2023] Open
Abstract
Polyglutamine (polyQ) diseases represent a group of progressive neurodegenerative disorders that are caused by abnormal expansion of CAG triplet nucleotides in disease genes. Recent evidence indicates that not only mutant polyQ proteins, but also their corresponding mutant RNAs, contribute to the pathogenesis of polyQ diseases. Here, we describe the identification of a 13-amino-acid peptide, P3, which binds directly and preferentially to long-CAG RNA within the pathogenic range. When administered to cell and Drosophila disease models, as well as to patient-derived fibroblasts, P3 inhibited expanded-CAG-RNA-induced nucleolar stress and suppressed neurotoxicity. We further examined the combined therapeutic effect of P3 and polyQ-binding peptide 1 (QBP1), a well-characterized polyQ protein toxicity inhibitor, on neurodegeneration. When P3 and QBP1 were co-administered to disease models, both RNA and protein toxicities were effectively mitigated, resulting in a notable improvement of neurotoxicity suppression compared with the P3 and QBP1 single-treatment controls. Our findings indicate that targeting toxic RNAs and/or simultaneous targeting of toxic RNAs and their corresponding proteins could open up a new therapeutic strategy for treating polyQ degeneration.
Collapse
Affiliation(s)
- Qian Zhang
- Laboratory of Drosophila Research, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Biochemistry Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Ho Tsoi
- Laboratory of Drosophila Research, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Biochemistry Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Shaohong Peng
- Laboratory of Drosophila Research, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Biochemistry Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Pan P Li
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Program of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kwok-Fai Lau
- Biochemistry Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Cell and Molecular Biology Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Molecular Biotechnology Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Dobrila D Rudnicki
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Program of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jacky Chi-Ki Ngo
- Biochemistry Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Cell and Molecular Biology Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Ho Yin Edwin Chan
- Laboratory of Drosophila Research, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Biochemistry Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Cell and Molecular Biology Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Molecular Biotechnology Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| |
Collapse
|
18
|
De Genst E, Chirgadze DY, Klein FAC, Butler DC, Matak-Vinković D, Trottier Y, Huston JS, Messer A, Dobson CM. Structure of a single-chain Fv bound to the 17 N-terminal residues of huntingtin provides insights into pathogenic amyloid formation and suppression. J Mol Biol 2015; 427:2166-78. [PMID: 25861763 PMCID: PMC4451460 DOI: 10.1016/j.jmb.2015.03.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/17/2015] [Accepted: 03/30/2015] [Indexed: 10/25/2022]
Abstract
Huntington's disease is triggered by misfolding of fragments of mutant forms of the huntingtin protein (mHTT) with aberrant polyglutamine expansions. The C4 single-chain Fv antibody (scFv) binds to the first 17 residues of huntingtin [HTT(1-17)] and generates substantial protection against multiple phenotypic pathologies in situ and in vivo. We show in this paper that C4 scFv inhibits amyloid formation by exon1 fragments of huntingtin in vitro and elucidate the structural basis for this inhibition and protection by determining the crystal structure of the complex of C4 scFv and HTT(1-17). The peptide binds with residues 3-11 forming an amphipathic helix that makes contact with the antibody fragment in such a way that the hydrophobic face of this helix is shielded from the solvent. Residues 12-17 of the peptide are in an extended conformation and interact with the same region of another C4 scFv:HTT(1-17) complex in the asymmetric unit, resulting in a β-sheet interface within a dimeric C4 scFv:HTT(1-17) complex. The nature of this scFv-peptide complex was further explored in solution by high-resolution NMR and physicochemical analysis of species in solution. The results provide insights into the manner in which C4 scFv inhibits the aggregation of HTT, and hence into its therapeutic potential, and suggests a structural basis for the initial interactions that underlie the formation of disease-associated amyloid fibrils by HTT.
Collapse
Affiliation(s)
- Erwin De Genst
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Dimitri Y Chirgadze
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Fabrice A C Klein
- Translational Medicine and Neurogenetics Programme, Institute of Genetics and Molecular and Cellular Biology, 67404 Illkirch Cédex, France
| | - David C Butler
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA; Department of Biomedical Sciences, University at Albany, Albany, NY 12208, USA
| | - Dijana Matak-Vinković
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Yvon Trottier
- Translational Medicine and Neurogenetics Programme, Institute of Genetics and Molecular and Cellular Biology, 67404 Illkirch Cédex, France
| | - James S Huston
- James S. Huston, The Antibody Society, Newton, MA 02462, USA
| | - Anne Messer
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA; Department of Biomedical Sciences, University at Albany, Albany, NY 12208, USA
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
19
|
Hoffner G, Djian P. Polyglutamine Aggregation in Huntington Disease: Does Structure Determine Toxicity? Mol Neurobiol 2014; 52:1297-1314. [PMID: 25336039 DOI: 10.1007/s12035-014-8932-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/09/2014] [Indexed: 01/14/2023]
Abstract
Huntington disease is a dominantly inherited disease of the central nervous system. The mutational expansion of polyglutamine beyond a critical length produces a toxic gain of function in huntingtin and results in neuronal death. In the course of the disease, expanded huntingtin is proteolyzed, becomes abnormally folded, and accumulates in oligomers, fibrils, and microscopic inclusions. The aggregated forms of the expanded protein are structurally diverse. Structural heterogeneity may explain why polyglutamine-containing aggregates could paradoxically be either toxic or neuroprotective. When defined, the toxic structures could then specifically be targeted by prophylactic or therapeutic drugs aimed at inhibiting polyglutamine aggregation.
Collapse
Affiliation(s)
- Guylaine Hoffner
- Laboratoire de Physiologie Cérébrale, Centre National de la Recherche Scientifique, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France
| | - Philippe Djian
- Laboratoire de Physiologie Cérébrale, Centre National de la Recherche Scientifique, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| |
Collapse
|
20
|
Schapira AHV, Olanow CW, Greenamyre JT, Bezard E. Slowing of neurodegeneration in Parkinson's disease and Huntington's disease: future therapeutic perspectives. Lancet 2014; 384:545-55. [PMID: 24954676 DOI: 10.1016/s0140-6736(14)61010-2] [Citation(s) in RCA: 287] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several important advances have been made in our understanding of the pathways that lead to cell dysfunction and death in Parkinson's disease and Huntington's disease. These advances have been informed by both direct analysis of the post-mortem brain and by study of the biological consequences of the genetic causes of these diseases. Some of the pathways that have been implicated so far include mitochondrial dysfunction, oxidative stress, kinase pathways, calcium dysregulation, inflammation, protein handling, and prion-like processes. Intriguingly, these pathways seem to be important in the pathogenesis of both diseases and have led to the identification of molecular targets for candidate interventions designed to slow or reverse their course. We review some recent advances that underlie putative therapies for neuroprotection in Parkinson's disease and Huntington's disease, and potential targets that might be exploited in the future. Although we will need to overcome important hurdles, especially in terms of clinical trial design, we propose several target pathways that merit further study. In Parkinson's disease, these targets include agents that might improve mitochondrial function or increase degradation of defective mitochondria, kinase inhibitors, calcium channel blockers, and approaches that interfere with the misfolding, templating, and transmission of α-synuclein. In Huntington's disease, strategies might also be directed at mitochondrial bioenergetics and turnover, the prevention of protein dysregulation, disruption of the interaction between huntingtin and p53 or huntingtin-interacting protein 1 to reduce apoptosis, and interference with expression of mutant huntingtin at both the nucleic acid and protein levels.
Collapse
Affiliation(s)
| | - C Warren Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, 33000 Bordeaux, France
| |
Collapse
|
21
|
Todd TW, Lim J. Aggregation formation in the polyglutamine diseases: protection at a cost? Mol Cells 2013; 36:185-94. [PMID: 23794019 PMCID: PMC3800151 DOI: 10.1007/s10059-013-0167-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/02/2013] [Indexed: 12/30/2022] Open
Abstract
Mutant protein aggregation is a hallmark of many neurodegenerative diseases, including the polyglutamine disorders. Although the correlation between aggregation formation and disease pathology originally suggested that the visible inclusions seen in patient tissue might directly contribute to pathology, additional studies failed to confirm this hypothesis. Current opinion in the field of polyglutamine disease research now favors a model in which large inclusions are cytoprotective and smaller oligomers or misfolded monomers underlie pathogenesis. Nonetheless, therapies aimed at reducing or preventing aggregation show promise. This review outlines the debate about the role of aggregation in the polyglutamine diseases as it has unfolded in the literature and concludes with a brief discussion on the manipulation of aggregation formation and clearance mechanisms as a means of therapeutic intervention.
Collapse
Affiliation(s)
- Tiffany W. Todd
- Department of Genetics, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Janghoo Lim
- Department of Genetics, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
22
|
Inhibiting toxic aggregation of amyloidogenic proteins: a therapeutic strategy for protein misfolding diseases. Biochim Biophys Acta Gen Subj 2013; 1830:4860-71. [PMID: 23820032 DOI: 10.1016/j.bbagen.2013.06.029] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND The deposition of self-assembled amyloidogenic proteins is associated with multiple diseases, including Alzheimer's disease, Parkinson's disease and type 2 diabetes mellitus. The toxic misfolding and self-assembling of amyloidogenic proteins are believed to underlie protein misfolding diseases. Novel drug candidates targeting self-assembled amyloidogenic proteins represent a potential therapeutic approach for protein misfolding diseases. SCOPE OF REVIEW In this perspective review, we provide an overview of the recent progress in identifying inhibitors that block the aggregation of amyloidogenic proteins and the clinical applications thereof. MAJOR CONCLUSIONS Compounds such as polyphenols, certain short peptides, and monomer- or oligomer-specific antibodies, can interfere with the self-assembly of amyloidogenic proteins, prevent the formation of oligomers, amyloid fibrils and the consequent cytotoxicity. GENERAL SIGNIFICANCE Some inhibitors have been tested in clinical trials for treating protein misfolding diseases. Inhibitors that target the aggregation of amyloidogenic proteins bring new hope to therapy for protein misfolding diseases.
Collapse
|
23
|
Margulis BA, Vigont V, Lazarev VF, Kaznacheyeva EV, Guzhova IV. Pharmacological protein targets in polyglutamine diseases: mutant polypeptides and their interactors. FEBS Lett 2013; 587:1997-2007. [PMID: 23684638 DOI: 10.1016/j.febslet.2013.05.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 12/18/2022]
Abstract
Polyglutamine diseases are a group of pathologies affecting different parts of the brain and causing dysfunction and atrophy of certain neural cell populations. These diseases stem from mutations in various cellular genes that result in the synthesis of proteins with extended polyglutamine tracts. In particular, this concerns huntingtin, ataxins, and androgen receptor. These mutant proteins can form oligomers, aggregates, and, finally, aggresomes with distinct functions and different degrees of cytotoxicity. In this review, we analyze the effects of different forms of polyQ proteins on other proteins and their functions, which are considered as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Boris A Margulis
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky pr., 4, St. Petersburg 194064, Russia
| | | | | | | | | |
Collapse
|