1
|
Riessland M, Ximerakis M, Jarjour AA, Zhang B, Orr ME. Therapeutic targeting of senescent cells in the CNS. Nat Rev Drug Discov 2024; 23:817-837. [PMID: 39349637 DOI: 10.1038/s41573-024-01033-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 11/01/2024]
Abstract
Senescent cells accumulate throughout the body with advanced age, diseases and chronic conditions. They negatively impact health and function of multiple systems, including the central nervous system (CNS). Therapies that target senescent cells, broadly referred to as senotherapeutics, recently emerged as potentially important treatment strategies for the CNS. Promising therapeutic approaches involve clearing senescent cells by disarming their pro-survival pathways with 'senolytics'; or dampening their toxic senescence-associated secretory phenotype (SASP) using 'senomorphics'. Following the pioneering discovery of first-generation senolytics dasatinib and quercetin, dozens of additional therapies have been identified, and several promising targets are under investigation. Although potentially transformative, senotherapies are still in early stages and require thorough testing to ensure reliable target engagement, specificity, safety and efficacy. The limited brain penetrance and potential toxic side effects of CNS-acting senotherapeutics pose challenges for drug development and translation to the clinic. This Review assesses the potential impact of senotherapeutics for neurological conditions by summarizing preclinical evidence, innovative methods for target and biomarker identification, academic and industry drug development pipelines and progress in clinical trials.
Collapse
Affiliation(s)
- Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | | | | | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miranda E Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Salisbury VA Medical Center, Salisbury, NC, USA.
| |
Collapse
|
2
|
Ahanger IA, Dar TA. Small molecule modulators of alpha-synuclein aggregation and toxicity: Pioneering an emerging arsenal against Parkinson's disease. Ageing Res Rev 2024; 101:102538. [PMID: 39389237 DOI: 10.1016/j.arr.2024.102538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Parkinson's disease (PD) is primarily characterized by loss of dopaminergic neurons in the substantia nigra pars compacta region of the brain and accumulation of aggregated forms of alpha-synuclein (α-Syn), an intrinsically disordered protein, in the form of Lewy Bodies and Lewy Neurites. Substantial evidences point to the aggregated/fibrillar forms of α-Syn as a central event in PD pathogenesis, underscoring the modulation of α-Syn aggregation as a promising strategy for PD treatment. Consequently, numerous anti-aggregation agents, spanning from small molecules to polymers, have been scrutinized for their potential to mitigate α-Syn aggregation and its associated toxicity. Among these, small molecule modulators like osmoprotectants, polyphenols, cellular metabolites, metals, and peptides have emerged as promising candidates with significant potential in PD management. This article offers a comprehensive overview of the effects of these small molecule modulators on the aggregation propensity and associated toxicity of α-Syn and its PD-associated mutants. It serves as a valuable resource for identifying and developing potent, non-invasive, non-toxic, and highly specific small molecule-based therapeutic arsenal for combating PD. Additionally, it raises pertinent questions aimed at guiding future research endeavours in the field of α-Syn aggregation remodelling.
Collapse
Affiliation(s)
- Ishfaq Ahmad Ahanger
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| |
Collapse
|
3
|
Kitson RRA, Kitsonová D, Siegel D, Ross D, Moody CJ. Geldanamycin, a Naturally Occurring Inhibitor of Hsp90 and a Lead Compound for Medicinal Chemistry. J Med Chem 2024; 67:17946-17963. [PMID: 39361055 PMCID: PMC11513894 DOI: 10.1021/acs.jmedchem.4c01048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Geldanamycin remains a driver in the medicinal chemistry of heat shock protein 90 (Hsp90) inhibition, even half a century after its original isolation from nature. This Perspective focuses on the properties of the benzoquinone ring of the natural product that enable a range of functionalization reactions to take place. Therefore, inherent reactivity at C-17, where the methoxy group serves as a vinylogous ester, and at C-19 that demonstrates nucleophilic, enamide-type character toward electrophiles, and also as a conjugate acceptor to react with nucleophiles, has facilitated the synthesis of semisynthetic derivatives. Thus, a range of C-17-substituted amine derivatives has been investigated in oncology applications, with a number of compounds in this series reaching clinical trials. In contrast, the 19-position of geldanamycin has received less attention, although 19-substituted derivatives offer promise with markedly reduced toxicity compared to geldanamycin itself, while retaining Hsp90 inhibitory activity albeit with diminished potency in cellular studies.
Collapse
Affiliation(s)
- Russell R. A. Kitson
- Department
of Organic and Bioorganic Chemistry, Charles
University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Dominika Kitsonová
- Datwyler
Sealing Technologies CZ Ltd., Polní 224, 50401 Nový Bydžov, Czech
Republic
| | - David Siegel
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - David Ross
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - Christopher J. Moody
- School
of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| |
Collapse
|
4
|
Sharma A, Shah OP, Sharma L, Gulati M, Behl T, Khalid A, Mohan S, Najmi A, Zoghebi K. Molecular Chaperones as Therapeutic Target: Hallmark of Neurodegenerative Disorders. Mol Neurobiol 2024; 61:4750-4767. [PMID: 38127187 DOI: 10.1007/s12035-023-03846-2] [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: 09/12/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
Misfolded and aggregated proteins build up in neurodegenerative illnesses, which causes neuronal dysfunction and ultimately neuronal death. In the last few years, there has been a significant upsurge in the level of interest towards the function of molecular chaperones in the control of misfolding and aggregation. The crucial molecular chaperones implicated in neurodegenerative illnesses are covered in this review article, along with a variety of their different methods of action. By aiding in protein folding, avoiding misfolding, and enabling protein breakdown, molecular chaperones serve critical roles in preserving protein homeostasis. By aiding in protein folding, avoiding misfolding, and enabling protein breakdown, molecular chaperones have integral roles in preserving regulation of protein balance. It has been demonstrated that aging, a significant risk factor for neurological disorders, affects how molecular chaperones function. The aggregation of misfolded proteins and the development of neurodegeneration may be facilitated by the aging-related reduction in chaperone activity. Molecular chaperones have also been linked to the pathophysiology of several instances of neuron withering illnesses, enumerating as Parkinson's disease, Huntington's disease, and Alzheimer's disease. Molecular chaperones have become potential therapy targets concerning with the prevention and therapeutic approach for brain disorders due to their crucial function in protein homeostasis and their connection to neurodegenerative illnesses. Protein homeostasis can be restored, and illness progression can be slowed down by methods that increase chaperone function or modify their expression. This review emphasizes the importance of molecular chaperones in the context of neuron withering disorders and their potential as therapeutic targets for brain disorders.
Collapse
Affiliation(s)
- Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Om Prakash Shah
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 1444411, India
- ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW, 20227, Australia
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India, Amity University, Mohali, India.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, 45142, Saudi Arabia
- Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box 2424, 11111, Khartoum, Sudan
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, 45142, Saudi Arabia.
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| |
Collapse
|
5
|
Schwab K, Frahm S, Magbagbeolu M, Horsley D, Goatman EA, Melis V, Theuring F, Ishaq A, Storey JMD, Harrington CR, Wischik CM, Riedel G. LETC inhibits α-Syn aggregation and ameliorates motor deficiencies in the L62 mouse model of synucleinopathy. Eur J Pharmacol 2024; 970:176505. [PMID: 38503400 DOI: 10.1016/j.ejphar.2024.176505] [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: 01/16/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024]
Abstract
Alpha-Synuclein (α-Syn) aggregation is a pathological feature of synucleinopathies, neurodegenerative disorders that include Parkinson's disease (PD). Here, we explored the efficacy of N,N,N',N'-tetraethyl-10H-phenothiazine-3,7-diamine dihydrochloride (LETC), a protein aggregation inhibitor, on α-Syn aggregation. In both cellular models and transgenic mice, α-Syn aggregation was achieved by the overexpression of full-length human α-Syn fused with a signal sequence peptide. α-Syn accumulated in transfected DH60.21 neuroblastoma cells and α-Syn aggregation was inhibited by LETC with an EC50 of 0.066 ± 0.047 μM. Full-length human α-Syn overexpressing Line 62 (L62) mice accumulated neuronal α-Syn that was associated with a decreased motor performance in the open field and automated home cage. LETC, administered orally for 6 weeks at 10 mg/kg significantly decreased α-Syn-positive neurons in multiple brain regions and this resulted in a rescue of movement deficits in the open field in these mice. LETC however, did not improve activity deficits of L62 mice in the home cage environment. The results suggest that LETC may provide a potential disease modification therapy in synucleinopathies through the inhibition of α-Syn aggregation.
Collapse
Affiliation(s)
- Karima Schwab
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany.
| | - Silke Frahm
- Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Mandy Magbagbeolu
- Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - David Horsley
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Elizabeth A Goatman
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Valeria Melis
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Franz Theuring
- Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Ahtsham Ishaq
- Department of Chemistry, University of Aberdeen, Aberdeen, UK
| | - John M D Storey
- Department of Chemistry, University of Aberdeen, Aberdeen, UK; TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Charles R Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Claude M Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
6
|
Kinger S, Jagtap YA, Kumar P, Choudhary A, Prasad A, Prajapati VK, Kumar A, Mehta G, Mishra A. Proteostasis in neurodegenerative diseases. Adv Clin Chem 2024; 121:270-333. [PMID: 38797543 DOI: 10.1016/bs.acc.2024.04.002] [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: 05/29/2024]
Abstract
Proteostasis is essential for normal function of proteins and vital for cellular health and survival. Proteostasis encompasses all stages in the "life" of a protein, that is, from translation to functional performance and, ultimately, to degradation. Proteins need native conformations for function and in the presence of multiple types of stress, their misfolding and aggregation can occur. A coordinated network of proteins is at the core of proteostasis in cells. Among these, chaperones are required for maintaining the integrity of protein conformations by preventing misfolding and aggregation and guide those with abnormal conformation to degradation. The ubiquitin-proteasome system (UPS) and autophagy are major cellular pathways for degrading proteins. Although failure or decreased functioning of components of this network can lead to proteotoxicity and disease, like neuron degenerative diseases, underlying factors are not completely understood. Accumulating misfolded and aggregated proteins are considered major pathomechanisms of neurodegeneration. In this chapter, we have described the components of three major branches required for proteostasis-chaperones, UPS and autophagy, the mechanistic basis of their function, and their potential for protection against various neurodegenerative conditions, like Alzheimer's, Parkinson's, and Huntington's disease. The modulation of various proteostasis network proteins, like chaperones, E3 ubiquitin ligases, proteasome, and autophagy-associated proteins as therapeutic targets by small molecules as well as new and unconventional approaches, shows promise.
Collapse
Affiliation(s)
- Sumit Kinger
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Yuvraj Anandrao Jagtap
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Prashant Kumar
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Akash Choudhary
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Amit Prasad
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Dhaula Kuan, New Delhi, India
| | - Amit Kumar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, India
| | - Gunjan Mehta
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Telangana, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India.
| |
Collapse
|
7
|
Mansour HM, Mohamed AF, Khattab MM, El-Khatib AS. Heat Shock Protein 90 in Parkinson's Disease: Profile of a Serial Killer. Neuroscience 2024; 537:32-46. [PMID: 38040085 DOI: 10.1016/j.neuroscience.2023.11.031] [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: 09/29/2023] [Revised: 11/18/2023] [Accepted: 11/26/2023] [Indexed: 12/03/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, characterized by abnormal α-synuclein misfolding and aggregation, mitochondrial dysfunction, oxidative stress, as well as progressive death of dopaminergic neurons in the substantia nigra. Molecular chaperones play a role in stabilizing proteins and helping them achieve their proper structure. Previous studies have shown that overexpression of heat shock protein 90 (HSP90) can lead to the death of dopaminergic neurons associated with PD. Inhibiting HSP90 is considered a potential treatment approach for neurodegenerative disorders, as it may reduce protein aggregation and related toxicity, as well as suppress various forms of regulated cell death (RCD). This review provides an overview of HSP90 and its role in PD, focusing on its modulation of proteostasis and quality control of LRRK2. The review also explores the effects of HSP90 on different types of RCD, such as apoptosis, chaperone-mediated autophagy (CMA), necroptosis, and ferroptosis. Additionally, it discusses HSP90 inhibitors that have been tested in PD models. We will highlight the under-investigated neuroprotective effects of HSP90 inhibition, including modulation of oxidative stress, mitochondrial dysfunction, PINK/PARKIN, heat shock factor 1 (HSF1), histone deacetylase 6 (HDAC6), and the PHD2-HSP90 complex-mediated mitochondrial stress pathway. By examining previous literature, this review uncovers overlooked neuroprotective mechanisms and emphasizes the need for further research on HSP90 inhibitors as potential therapeutic strategies for PD. Finally, the review discusses the potential limitations and possibilities of using HSP90 inhibitors in PD therapy.
Collapse
Affiliation(s)
- Heba M Mansour
- Central Administration of Biological, Innovative Products, and Clinical Studies (BIO-INN), Egyptian Drug Authority, EDA, Giza, Egypt.
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Bastian P, Konieczna L, Dulski J, Daca A, Jacewicz D, Płoska A, Knap N, Sławek J, Bączek T, Kalinowski L, Drzeżdżon J, Roszmann A, Belka M, Górska-Ponikowska M. 2-Methoxyestradiol and Hydrogen Peroxide as Promising Biomarkers in Parkinson's Disease. Mol Neurobiol 2024; 61:148-166. [PMID: 37589832 PMCID: PMC10791893 DOI: 10.1007/s12035-023-03575-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Estrogens function in numerous physiological processes including controlling brain cell growth and differentiation. 2-Methoxestradiol (2-ME2), a 17β-estradiol (E2) metabolite, is known for its anticancer effects as observed both in vivo and in vitro. 2-ME2 affects all actively dividing cells, including neurons. The study aimed to determine whether 2-ME2 is a potentially cancer-protective or rather neurodegenerative agent in a specific tissue culture model as well as a clinical setup. In this study, 2-ME2 activity was determined in a Parkinson's disease (PD) in vitro model based on the neuroblastoma SH-SY5Y cell line. The obtained results suggest that 2-ME2 generates nitro-oxidative stress and controls heat shock proteins (HSP), resulting in DNA strand breakage and apoptosis. On the one hand, it may affect intensely dividing cells preventing cancer development; however, on the other hand, this kind of activity within the central nervous system may promote neurodegenerative diseases like PD. Thus, the translational value of 2-ME2's neurotoxic activity in a PD in vitro model was also investigated. LC-MS/MS technique was used to evaluate estrogens and their derivatives, namely, hydroxy and methoxyestrogens, in PD patients' blood, whereas the stopped-flow method was used to assess hydrogen peroxide (H2O2) levels. Methoxyestrogens and H2O2 levels were increased in patients' blood as compared to control subjects, but hydoxyestrogens were simultaneously decreased. From the above, we suggest that the determination of plasma levels of methoxyestrogens and H2O2 may be a novel PD biomarker. The presented research is the subject of the pending patent application "The use of hydrogen peroxide and 17β-estradiol and its metabolites as biomarkers in the diagnosis of neurodegenerative diseases," no. P.441360.
Collapse
Affiliation(s)
- Paulina Bastian
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland.
| | - Lucyna Konieczna
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Jarosław Dulski
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Dagmara Jacewicz
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Jarosław Sławek
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211, Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza Street 11/12, 80-233, Gdansk, Poland
| | - Joanna Drzeżdżon
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Anna Roszmann
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Magdalena Górska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569, Stuttgart, Germany.
- Euro-Mediterranean Institute of Science and Technology, 90139, Palermo, Italy.
| |
Collapse
|
9
|
Stylianopoulou E, Daviti A, Giourou V, Gerasimidi E, Nikolaou A, Kourkoutas Y, Grigoriou ME, Paleologou KE, Skavdis G. Assessment of the Anti-Amyloidogenic Properties of Essential Oils and Their Constituents in Cells Using a Whole-Cell Recombinant Biosensor. Brain Sci 2023; 14:35. [PMID: 38248250 PMCID: PMC10812981 DOI: 10.3390/brainsci14010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Essential oils exhibit numerous medicinal properties, including antimicrobial, anti-inflammatory and antioxidant effects. Recent studies also indicate that certain essential oils demonstrate anti-amyloidogenic activity against β-amyloid, the protein implicated in Alzheimer's disease. To investigate whether the anti-aggregating properties of essential oils extend to α-synuclein, the protein involved in Parkinson's disease, we constructed and employed a whole-cell biosensor based on the split-luciferase complementation assay. We validated our biosensor by using baicalein, a known inhibitor of α-synuclein aggregation, and subsequently we tested eight essential oils commonly used in food and the hygienic industry. Two of them, citron and sage, along with their primary components, pure linalool (the main constituent in citron essential oil) and pure eucalyptol (1,8-cineole, the main constituent in sage essential oil), were able to reduce α-syn aggregation. These findings suggest that both essential oils and their main constituents could be regarded as potential components in functional foods or incorporated into complementary Parkinson's disease therapies.
Collapse
Affiliation(s)
- Electra Stylianopoulou
- Laboratory of Developmental Biology & Molecular Neurobiology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.S.); (A.D.); (E.G.); (M.E.G.)
- Laboratory of Molecular Regulation & Diagnostic Technology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Anastasia Daviti
- Laboratory of Developmental Biology & Molecular Neurobiology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.S.); (A.D.); (E.G.); (M.E.G.)
| | - Venetia Giourou
- Laboratory of Molecular Regulation & Diagnostic Technology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Eleni Gerasimidi
- Laboratory of Developmental Biology & Molecular Neurobiology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.S.); (A.D.); (E.G.); (M.E.G.)
| | - Anastasios Nikolaou
- Laboratory of Applied Microbiology & Biotechnology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.N.); (Y.K.)
| | - Yiannis Kourkoutas
- Laboratory of Applied Microbiology & Biotechnology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.N.); (Y.K.)
| | - Maria E. Grigoriou
- Laboratory of Developmental Biology & Molecular Neurobiology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.S.); (A.D.); (E.G.); (M.E.G.)
| | - Katerina E. Paleologou
- Laboratory of Developmental Biology & Molecular Neurobiology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.S.); (A.D.); (E.G.); (M.E.G.)
| | - George Skavdis
- Laboratory of Molecular Regulation & Diagnostic Technology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
10
|
Silvestro S, Raffaele I, Mazzon E. Modulating Stress Proteins in Response to Therapeutic Interventions for Parkinson's Disease. Int J Mol Sci 2023; 24:16233. [PMID: 38003423 PMCID: PMC10671288 DOI: 10.3390/ijms242216233] [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: 10/03/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative illness characterized by the degeneration of dopaminergic neurons in the substantia nigra, resulting in motor symptoms and without debilitating motors. A hallmark of this condition is the accumulation of misfolded proteins, a phenomenon that drives disease progression. In this regard, heat shock proteins (HSPs) play a central role in the cellular response to stress, shielding cells from damage induced by protein aggregates and oxidative stress. As a result, researchers have become increasingly interested in modulating these proteins through pharmacological and non-pharmacological therapeutic interventions. This review aims to provide an overview of the preclinical experiments performed over the last decade in this research field. Specifically, it focuses on preclinical studies that center on the modulation of stress proteins for the treatment potential of PD. The findings display promise in targeting HSPs to ameliorate PD outcomes. Despite the complexity of HSPs and their co-chaperones, proteins such as HSP70, HSP27, HSP90, and glucose-regulated protein-78 (GRP78) may be efficacious in slowing or preventing disease progression. Nevertheless, clinical validation is essential to confirm the safety and effectiveness of these preclinical approaches.
Collapse
Affiliation(s)
| | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (I.R.)
| |
Collapse
|
11
|
Basu S, Song M, Adams L, Jeong I, Je G, Guhathakurta S, Jiang J, Boparai N, Dai W, Cardozo-Pelaez F, Tatulian SA, Han KY, Elliott J, Baum J, McLean PJ, Dickson DW, Kim YS. Transcriptional mutagenesis of α-synuclein caused by DNA oxidation in Parkinson's disease pathogenesis. Acta Neuropathol 2023; 146:685-705. [PMID: 37740734 PMCID: PMC10564827 DOI: 10.1007/s00401-023-02632-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/09/2023] [Accepted: 09/10/2023] [Indexed: 09/25/2023]
Abstract
Oxidative stress plays an essential role in the development of Parkinson's disease (PD). 8-oxo-7,8-dihydroguanine (8-oxodG, oxidized guanine) is the most abundant oxidative stress-mediated DNA lesion. However, its contributing role in underlying PD pathogenesis remains unknown. In this study, we hypothesized that 8-oxodG can generate novel α-synuclein (α-SYN) mutants with altered pathologic aggregation through a phenomenon called transcriptional mutagenesis (TM). We observed a significantly higher accumulation of 8-oxodG in the midbrain genomic DNA from PD patients compared to age-matched controls, both globally and region specifically to α-SYN. In-silico analysis predicted that forty-three amino acid positions can contribute to TM-derived α-SYN mutation. Here, we report a significantly higher load of TM-derived α-SYN mutants from the midbrain of PD patients compared to controls using a sensitive PCR-based technique. We found a novel Serine42Tyrosine (S42Y) α-SYN as the most frequently detected TM mutant, which incidentally had the highest predicted aggregation score amongst all TM variants. Immunohistochemistry of midbrain sections from PD patients using a newly characterized antibody for S42Y identified S42Y-laden Lewy bodies (LB). We further demonstrated that the S42Y TM variant significantly accelerates WT α-SYN aggregation by cell and recombinant protein-based assays. Cryo-electron tomography revealed that S42Y exhibits considerable conformational heterogeneity compared to WT fibrils. Moreover, S42Y exhibited higher neurotoxicity compared to WT α-SYN as shown in mouse primary cortical cultures and AAV-mediated overexpression in the substantia nigra of C57BL/6 J mice. To our knowledge, this is the first report describing the possible contribution of TM-generated mutations of α-SYN to LB formation and PD pathogenesis.
Collapse
Affiliation(s)
- Sambuddha Basu
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Minkyung Song
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Levi Adams
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Inhye Jeong
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Goun Je
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Subhrangshu Guhathakurta
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Jennifer Jiang
- Department of Cell Biology and Neuroscience, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ, 08854, USA
| | - Nikpreet Boparai
- Department of Cell Biology and Neuroscience, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ, 08854, USA
| | - Wei Dai
- Department of Cell Biology and Neuroscience, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ, 08854, USA
| | - Fernando Cardozo-Pelaez
- Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
- Center for Structural and Functional Neurosciences, University of Montana, Missoula, MT, 59812, USA
| | - Suren A Tatulian
- Department of Physics, University of Central Florida, Orlando, FL, 32816, USA
| | - Kyu Young Han
- CREOL, The College of Optics and Photonics, University of Central Florida, Orlando, FL, USA
| | - Jordan Elliott
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Yoon-Seong Kim
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA.
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, 32827, USA.
| |
Collapse
|
12
|
Mohammed S, Russo I, Ramazzina I. Uncovering the Role of Natural and Synthetic Small Molecules in Counteracting the Burden of α-Synuclein Aggregates and Related Toxicity in Different Models of Parkinson's Disease. Int J Mol Sci 2023; 24:13370. [PMID: 37686175 PMCID: PMC10488152 DOI: 10.3390/ijms241713370] [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: 08/02/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
A proteostasis network represents a sophisticated cellular system that controls the whole process which leads to properly folded functional proteins. The imbalance of proteostasis determines a quantitative increase in misfolded proteins prone to aggregation and elicits the onset of different diseases. Among these, Parkinson's Disease (PD) is a progressive brain disorder characterized by motor and non-motor signs. In PD pathogenesis, alpha-Synuclein (α-Syn) loses its native structure, triggering a polymerization cascade that leads to the formation of toxic inclusions, the PD hallmark. Because molecular chaperones represent a "cellular arsenal" to counteract protein misfolding and aggregation, the modulation of their expression represents a compelling PD therapeutic strategy. This review will discuss evidence concerning the effects of natural and synthetic small molecules in counteracting α-Syn aggregation process and related toxicity, in different in vitro and in vivo PD models. Firstly, the role of small molecules that modulate the function(s) of chaperones will be highlighted. Then, attention will be paid to small molecules that interfere with different steps of the protein-aggregation process. This overview would stimulate in-depth research on already-known small molecules or the development of new ones, with the aim of developing drugs that are able to modify the progression of the disease.
Collapse
Affiliation(s)
- Salihu Mohammed
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy;
| | - Isabella Russo
- Department of Molecular and Translational Medicine, University of Brescia, Via Europa 11, 25123 Brescia, Italy;
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Ileana Ramazzina
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy;
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy
- Biostructures and Biosystems National Institute (INBB), Viale Medaglie d’Oro 305, 00136 Rome, Italy
| |
Collapse
|
13
|
Moyano P, Sola E, Naval MV, Guerra-Menéndez L, Fernández MDLC, del Pino J. Neurodegenerative Proteinopathies Induced by Environmental Pollutants: Heat Shock Proteins and Proteasome as Promising Therapeutic Tools. Pharmaceutics 2023; 15:2048. [PMID: 37631262 PMCID: PMC10458078 DOI: 10.3390/pharmaceutics15082048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Environmental pollutants' (EPs) amount and diversity have increased in recent years due to anthropogenic activity. Several neurodegenerative diseases (NDs) are theorized to be related to EPs, as their incidence has increased in a similar way to human EPs exposure and they reproduce the main ND hallmarks. EPs induce several neurotoxic effects, including accumulation and gradual deposition of misfolded toxic proteins, producing neuronal malfunction and cell death. Cells possess different mechanisms to eliminate these toxic proteins, including heat shock proteins (HSPs) and the proteasome system. The accumulation and deleterious effects of toxic proteins are induced through HSPs and disruption of proteasome proteins' homeostatic function by exposure to EPs. A therapeutic approach has been proposed to reduce accumulation of toxic proteins through treatment with recombinant HSPs/proteasome or the use of compounds that increase their expression or activity. Our aim is to review the current literature on NDs related to EP exposure and their relationship with the disruption of the proteasome system and HSPs, as well as to discuss the toxic effects of dysfunction of HSPs and proteasome and the contradictory effects described in the literature. Lastly, we cover the therapeutic use of developed drugs and recombinant proteasome/HSPs to eliminate toxic proteins and prevent/treat EP-induced neurodegeneration.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Bothanic, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Lucia Guerra-Menéndez
- Department of Physiology, Medicine School, San Pablo CEU University, 28003 Madrid, Spain
| | - Maria De la Cabeza Fernández
- Department of Chemistry and Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
14
|
Siwecka N, Saramowicz K, Galita G, Rozpędek-Kamińska W, Majsterek I. Inhibition of Protein Aggregation and Endoplasmic Reticulum Stress as a Targeted Therapy for α-Synucleinopathy. Pharmaceutics 2023; 15:2051. [PMID: 37631265 PMCID: PMC10459316 DOI: 10.3390/pharmaceutics15082051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
α-synuclein (α-syn) is an intrinsically disordered protein abundant in the central nervous system. Physiologically, the protein regulates vesicle trafficking and neurotransmitter release in the presynaptic terminals. Pathologies related to misfolding and aggregation of α-syn are referred to as α-synucleinopathies, and they constitute a frequent cause of neurodegeneration. The most common α-synucleinopathy, Parkinson's disease (PD), is caused by abnormal accumulation of α-syn in the dopaminergic neurons of the midbrain. This results in protein overload, activation of endoplasmic reticulum (ER) stress, and, ultimately, neural cell apoptosis and neurodegeneration. To date, the available treatment options for PD are only symptomatic and rely on dopamine replacement therapy or palliative surgery. As the prevalence of PD has skyrocketed in recent years, there is a pending issue for development of new disease-modifying strategies. These include anti-aggregative agents that target α-syn directly (gene therapy, small molecules and immunization), indirectly (modulators of ER stress, oxidative stress and clearance pathways) or combine both actions (natural compounds). Herein, we provide an overview on the characteristic features of the structure and pathogenic mechanisms of α-syn that could be targeted with novel molecular-based therapies.
Collapse
Affiliation(s)
| | | | | | | | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (N.S.); (K.S.); (G.G.); (W.R.-K.)
| |
Collapse
|
15
|
Ramirez LM, Zweckstetter M. Molecular-level interplay between intrinsically disordered clients and Hsp90. Curr Opin Chem Biol 2023; 74:102304. [PMID: 37068388 DOI: 10.1016/j.cbpa.2023.102304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 04/19/2023]
Abstract
Proteostasis is maintained by a network of molecular chaperones, a prominent member of which is the 90-kilodalton heat shock protein Hsp90. The chaperone function of Hsp90 has been extensively reviewed previously, emphasizing its ATPase activity and remodeling of folded client proteins. Experimental evidence implicating Hsp90 in neurodegenerative diseases has bolstered interest in the noncanonical chaperoning of intrinsically disordered protein (IDPs), however the interplay between Hsp90 and its disordered clients remains poorly understood. In this review we describe recent advances that have contributed to our understanding of the intricate mechanisms characterizing Hsp90-mediated chaperoning of the IDPs tau and α-synuclein and survey emerging insights into the modulation of the chaperone-client interplay in the context of neurodegeneration.
Collapse
Affiliation(s)
- Lisa Marie Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Gӧttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Gӧttingen, Germany; Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Gӧttingen, Germany.
| |
Collapse
|
16
|
Franks H, Wang R, Li M, Wang B, Wildmann A, Ortyl T, O’Brien S, Young D, Liao FF, Sakata K. Heat shock factor HSF1 regulates BDNF gene promoters upon acute stress in the hippocampus, together with pCREB. J Neurochem 2023; 165:131-148. [PMID: 36227087 PMCID: PMC10097844 DOI: 10.1111/jnc.15707] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/29/2022]
Abstract
Heat shock factor 1 (HSF1) is a master stress-responsive transcriptional factor, protecting cells from death. However, its gene regulation in vivo in the brain in response to neuronal stimuli remains elusive. Here, we investigated its direct regulation of the brain-derived neurotrophic factor (BDNF) gene (Bdnf) in response to acute neuronal stress stimuli in the brain. The results of immunohistochemistry and chromatin immunoprecipitation quantitative PCR (ChIP-qPCR) showed that administration of kainic acid (a glutamate receptor agonist inducing excitotoxity) to young adult mice induced HSF1 nuclear translocation and its binding to multiple Bdnf promoters in the hippocampus. Footshock, a physical stressor used for learning, also induced HSF1 binding to selected Bdnf promoters I and IV. This is, to our knowledge, the first demonstration of HSF1 gene regulation in response to neuronal stimuli in the hippocampus in vivo. HSF1 binding sites (HSEs) in Bdnf promoters I and IV were also detected when immunoprecipitated by an antibody of phosphorylated (p)CREB (cAMP-responsive element-binding protein), suggesting their possible interplay in acute stress-induced Bdnf transcription. Interestingly, their promoter binding patterns differed by KA and footshock, suggesting that HSF1 and pCREB orchestrate to render fine-tuned promoter control depending on the types of stress. Further, HSF1 overexpression increased Bdnf promoter activity in a luciferase assay, while virus infection of constitutively active-form HSF1 increased levels of BDNF mRNA and protein in vitro in primary cultured neurons. These results indicated that HSF1 activation of Bdnf promoter was sufficient to induce BDNF expression. Taken together, these results suggest that HSF1 promoter-specific control of Bdnf gene regulation plays an important role in neuronal protection and plasticity in the hippocampus in response to acute stress, possibly interplaying with pCREB.
Collapse
Affiliation(s)
- Hunter Franks
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Ruishan Wang
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Mingqi Li
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Bin Wang
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Ashton Wildmann
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Tyler Ortyl
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Shannon O’Brien
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Deborah Young
- Department of Pharmacology & Clinical Pharmacology, The
University of Auckland, Auckland, New Zealand
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Kazuko Sakata
- Department of Pharmacology, University of Tennessee Health
Science Center, Memphis, TN, USA
| |
Collapse
|
17
|
Cools R, Vermeulen K, Narykina V, Leitao RCF, Bormans G. Radiosynthesis and preclinical evaluation of [ 11C]SNX-ab as an Hsp90α,β isoform-selective PET probe for in vivo brain and tumour imaging. EJNMMI Radiopharm Chem 2023; 8:2. [PMID: 36715827 PMCID: PMC9886718 DOI: 10.1186/s41181-023-00189-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The molecular chaperone, Hsp90, is a key player in the protein quality control system that maintains homeostasis under cellular stress conditions. It is a homodimer with ATP-dependent activity, and is a prominent member of the chaperone machinery that stabilizes, matures and (re)folds an extensive list of client proteins. Hsp90 occurs as four isoforms, cytosolic Hsp90α and Hsp90β, mitochondrial TRAP1 and Grp94 present in the endoplasmic reticulum. An aberrant role of Hsp90 has been attributed to several cancers and neurodegenerative disorders. Consequently, Hsp90 has emerged as an attractive therapeutic target. However, pan-Hsp90 inhibition often leads to detrimental dose-limiting toxicities. Novel strategies for Hsp90-targeted therapy intend to avoid this by using isoform-specific Hsp90 inhibition. In this respect, the radiosynthesis of carbon-11 labeled SNX-ab was developed and [11C]SNX-ab was evaluated as a Hsp90α,β isoform-selective PET probe, which could potentially allow to quantify in vivo Hsp90α,β expression. RESULTS [11C]SNX-ab was synthesized with excellent radiochemical yields of 45% and high radiochemical purity (> 98%). In vitro autoradiography studies on tissue slices of healthy mouse brain, mouse B16.F10 melanoma and U87 glioblastoma using homologous (SNX-ab, SNX-0723) and heterologous (Onalespib and PU-H71) Hsp90 inhibitors demonstrated only limited reduction of tracer binding, indicating that the binding of [11C]SNX-ab was not fully Hsp90-specific. Similarly, [11C]SNX-ab binding to U87 cells was not efficiently inhibited by Hsp90 inhibitors. Ex vivo biodistribution studies in healthy mice revealed limited brain exposure of [11C]SNX-ab and predominantly hepatobiliary clearance, which was confirmed by in vivo full-body dynamic µPET studies. CONCLUSION Our results suggest that [11C]SNX-ab is not an ideal probe for in vivo visualization and quantification of Hsp90α/β expression levels in tumour and brain. Future research in the development of next-generation Hsp90 isoform-selective PET tracers is warranted to dissect the role played by each isoform towards disease pathology and support the development of subtype-specific Hsp90 therapeutics.
Collapse
Affiliation(s)
- Romy Cools
- grid.5596.f0000 0001 0668 7884Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Koen Vermeulen
- grid.5596.f0000 0001 0668 7884Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium ,grid.8953.70000 0000 9332 3503NURA, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Valeria Narykina
- grid.511015.1Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Renan C. F. Leitao
- grid.5596.f0000 0001 0668 7884Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Guy Bormans
- grid.5596.f0000 0001 0668 7884Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
18
|
The Role of α-Synuclein in SNARE-mediated Synaptic Vesicle Fusion. J Mol Biol 2023; 435:167775. [PMID: 35931109 DOI: 10.1016/j.jmb.2022.167775] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
Neuronal communication depends on exquisitely regulated membrane fusion between synaptic vesicles and presynaptic neurons, which results in neurotransmitter release in precisely timed patterns. Presynaptic dysfunctions are known to occur prior to the onset of neurodegenerative diseases, including Parkinson's disease. Synaptic accumulation of α-synuclein (α-Syn) oligomers has been implicated in the pathway leading to such outcomes. α-Syn oligomers exert aberrant effects on presynaptic fusion machinery through their interactions with synaptic vesicles and proteins. Here, we summarize in vitro bulk and single-vesicle assays for investigating the functions of α-Syn monomers and oligomers in synaptic vesicle fusion and then discuss the current understanding of the roles of α-Syn monomers and oligomers in synaptic vesicle fusion. Finally, we suggest a new therapeutic avenue specifically targeting the mechanisms of α-Syn oligomer toxicity rather than the oligomer itself.
Collapse
|
19
|
Mitra S, Dash R, Munni YA, Selsi NJ, Akter N, Uddin MN, Mazumder K, Moon IS. Natural Products Targeting Hsp90 for a Concurrent Strategy in Glioblastoma and Neurodegeneration. Metabolites 2022; 12:1153. [PMID: 36422293 PMCID: PMC9697676 DOI: 10.3390/metabo12111153] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 09/16/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common aggressive, resistant, and invasive primary brain tumors that share neurodegenerative actions, resembling many neurodegenerative diseases. Although multiple conventional approaches, including chemoradiation, are more frequent in GBM therapy, these approaches are ineffective in extending the mean survival rate and are associated with various side effects, including neurodegeneration. This review proposes an alternative strategy for managing GBM and neurodegeneration by targeting heat shock protein 90 (Hsp90). Hsp90 is a well-known molecular chaperone that plays essential roles in maintaining and stabilizing protein folding to degradation in protein homeostasis and modulates signaling in cancer and neurodegeneration by regulating many client protein substrates. The therapeutic benefits of Hsp90 inhibition are well-known for several malignancies, and recent evidence highlights that Hsp90 inhibitors potentially inhibit the aggressiveness of GBM, increasing the sensitivity of conventional treatment and providing neuroprotection in various neurodegenerative diseases. Herein, the overview of Hsp90 modulation in GBM and neurodegeneration progress has been discussed with a summary of recent outcomes on Hsp90 inhibition in various GBM models and neurodegeneration. Particular emphasis is also given to natural Hsp90 inhibitors that have been evidenced to show dual protection in both GBM and neurodegeneration.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Nusrat Jahan Selsi
- Product Development Department, Popular Pharmaceuticals Ltd., Dhaka 1207, Bangladesh
| | - Nasrin Akter
- Department of Clinical Pharmacy and Molecular Pharmacology, East West University Bangladesh, Dhaka 1212, Bangladesh
| | - Md Nazim Uddin
- Department of Pharmacy, Southern University Bangladesh, Chittagong 4000, Bangladesh
| | - Kishor Mazumder
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
- School of Optometry and Vision Science, UNSW Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| |
Collapse
|
20
|
Aghazadeh N, Beilankouhi EAV, Fakhri F, Gargari MK, Bahari P, Moghadami A, Khodabandeh Z, Valilo M. Involvement of heat shock proteins and parkin/α-synuclein axis in Parkinson's disease. Mol Biol Rep 2022; 49:11061-11070. [PMID: 36097120 DOI: 10.1007/s11033-022-07900-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurological diseases, next only to Alzheimer's disease (AD) in terms of prevalence. It afflicts about 2-3% of individuals over 65 years old. The etiology of PD is unknown and several environmental and genetic factors are involved. From a pathological point of view, PD is characterized by the loss of dopaminergic neurons in the substantia nigra, which causes the abnormal accumulation of α-synuclein (α-syn) (a component of Lewy bodies), which subsequently interact with heat shock proteins (HSPs), leading to apoptosis. Apoptosis is a vital pathway for establishing homeostasis in body tissues, which is regulated by pro-apoptotic and anti-apoptotic factors. Recent findings have shown that HSPs, especially HSP27 and HSP70, play a pivotal role in regulating apoptosis by influencing the factors involved in the apoptosis pathway. Moreover, it has been reported that the expression of these HSPs in the nervous system is high. Apart from this finding, investigations have suggested that HSP27 and HSP70 (related to parkin) show a potent protective and anti-apoptotic impact against the damaging outcomes of mutant α-syn toxicity to nerve cells. Therefore, in this study, we aimed to investigate the relationship between these HSPs and apoptosis in patients with PD.
Collapse
Affiliation(s)
- Nina Aghazadeh
- Department of biology, Islamic Azad University, Tabriz, Iran
| | | | - Farima Fakhri
- Research Institute for Neuroscience, Kerman University of Medical Sciences, Kerman, Iran
| | - Morad Kohandel Gargari
- Faculty of Medicine, Imamreza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Bahari
- Department of Clinical Biochemistry, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Aliasghar Moghadami
- Department of Clinical Biochemistry and Medical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zhila Khodabandeh
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Mohammad Valilo
- Department of Clinical Biochemistry and Medical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Stykel MG, Ryan SD. Nitrosative stress in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:104. [PMID: 35953517 PMCID: PMC9372037 DOI: 10.1038/s41531-022-00370-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s Disease (PD) is a neurodegenerative disorder characterized, in part, by the loss of dopaminergic neurons within the nigral-striatal pathway. Multiple lines of evidence support a role for reactive nitrogen species (RNS) in degeneration of this pathway, specifically nitric oxide (NO). This review will focus on how RNS leads to loss of dopaminergic neurons in PD and whether RNS accumulation represents a central signal in the degenerative cascade. Herein, we provide an overview of how RNS accumulates in PD by considering the various cellular sources of RNS including nNOS, iNOS, nitrate, and nitrite reduction and describe evidence that these sources are upregulating RNS in PD. We document that over 1/3 of the proteins that deposit in Lewy Bodies, are post-translationally modified (S-nitrosylated) by RNS and provide a broad description of how this elicits deleterious effects in neurons. In doing so, we identify specific proteins that are modified by RNS in neurons which are implicated in PD pathogenesis, with an emphasis on exacerbation of synucleinopathy. How nitration of alpha-synuclein (aSyn) leads to aSyn misfolding and toxicity in PD models is outlined. Furthermore, we delineate how RNS modulates known PD-related phenotypes including axo-dendritic-, mitochondrial-, and dopamine-dysfunctions. Finally, we discuss successful outcomes of therapeutics that target S-nitrosylation of proteins in Parkinson’s Disease related clinical trials. In conclusion, we argue that targeting RNS may be of therapeutic benefit for people in early clinical stages of PD.
Collapse
Affiliation(s)
- Morgan G Stykel
- The Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON N1G 2W1, ON, Canada
| | - Scott D Ryan
- The Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON N1G 2W1, ON, Canada. .,Neurodegenerative Disease Center, Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
22
|
Thapa K, Khan H, Kanojia N, Singh TG, Kaur A, Kaur G. Therapeutic Insights on Ferroptosis in Parkinson's disease. Eur J Pharmacol 2022; 930:175133. [DOI: 10.1016/j.ejphar.2022.175133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022]
|
23
|
Azari A, Goodarzi A, Jafarkhani B, Eghbali M, Karimi Z, Hosseini Balef SS, Irannejad H. Novel molecular targets and mechanisms for neuroprotective modulation in neurodegenerative disorders. Cent Nerv Syst Agents Med Chem 2022; 22:88-107. [PMID: 35713146 DOI: 10.2174/1871524922666220616092132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neuronal death underlies the symptoms of several human neurological disorders, including Alzheimer's, Parkinson's and Huntington's diseases, and amyotrophic lateral sclerosis that their precise pathophysiology have not yet been elucidated. According to various studies the prohibition is the best therapy with neuroprotective approaches which are advanced and safe methods. METHODS This review summarizes some of the already-known and newly emerged neuroprotective targets and strategies that their experimental effects have been reported. Accordingly, literature was studied from 2000 to 2021 and appropriate articles were searched in Google Scholar and Scopus with the keywords given in the Keywords section of the current review. RESULTS Lewy bodies are the histopathologic characteristics of neurodegenerative disorders and are protein-rich intracellular deposits in which Alpha-Synuclein is its major protein. Alpha-Synuclein's toxic potential provides a compelling rationale for therapeutic strategies aimed at decreasing its burden in neuronal cells through numerous pathways including ubiquitin-proteasome system and autophagy-lysosome Pathway, proteolytic breakdown via cathepsin D, kallikrein-6 (neurosin), calpain-1 or MMP9, heat shock proteins, and proteolysis targeting chimera which consists of a target protein ligand and an E3 ubiquitin ligase (E3) followed by target protein ubiquitination (PROTACs). Other targets that have been noticed recently are the mutant huntingtin, tau proteins and glycogen synthase kinase 3β that their accumulation proceeds extensive neuronal damage and up to the minute approach such as Proteolysis Targeting Chimera promotes its degradation in cells. As various studies demonstrated that Mendelian gene mutations can result into the neurodegenerative diseases, additional target that has gained much interest is epigenetics such as mutation, phosphodiesterase, RNA binding proteins and Nuclear respiratory factor 1. CONCLUSION The novel molecular targets and new strategies compiled and introduced here can be used by scientists to design and discover more efficient small molecule drugs against the neurodegenerative diseases. And also the genes in which their mutations can lead to the α-synuclein aggregation or accumulation are discussed and considered a valuable information of epigenetics in dementia.
Collapse
Affiliation(s)
- Aala Azari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amin Goodarzi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Behrouz Jafarkhani
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Eghbali
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Karimi
- Department of Obstetrics & Gynecology, Imam Khomeini hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Sajad Hosseini Balef
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
24
|
Di Domenico F, Lanzillotta C. The disturbance of protein synthesis/degradation homeostasis is a common trait of age-related neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:49-87. [PMID: 36088079 DOI: 10.1016/bs.apcsb.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Protein homeostasis or "proteostasis" represent the process that regulates the balance of the intracellular functional and "healthy" proteins. Proteostasis is fundamental to preserve physiological metabolic processes in the cell and it allow to respond to any given stimulus as the expression of components of the proteostasis network is customized according to the proteomic demands of different cellular environments. In conditions that promote unfolding/misfolding of proteins chaperones act as signaling molecules inducing extreme measures to either fix the problem or destroy unfolded proteins. When the chaperone machinery fails under pathological insults unfolded proteins induce the endoplasmic reticulum (ER) stress activating the unfolded protein response (UPR) machinery. The activation of the UPR restores ER proteostasis primarily through the transcriptional remodeling of ER protein folding, trafficking, and degradation pathways, such as the ubiquitin proteasome system (UPS). If these mechanisms do not manage to clear the aberrant proteins, proteasome overload and become defective, and misfolded proteins may form aggregates thus extending the UPR mechanism. These aggregates are then attempted to be cleared by macroautophagy. Impaired proteostasis promote the accumulation of misfolded proteins that exacerbate the damage to chaperones, surveillance systems and/or degradative activities. Remarkably, the removal of toxic misfolded proteins is critical for all cells, but it is especially significant in neurons since these cannot be readily replaced. In neurons, the maintenance of efficient proteostasis is essential to healthy aging since the dysregulation of the proteostasis network can lead to neurodegenerative disease. Each of these brain pathologies is characterized by the repeated misfolding of one of more peculiar proteins, which evade both the protein folding machinery and cellular degradation mechanisms and begins to form aggregates that nucleate out into large fibrillar aggregates. In this chapter we describe the mechanisms, associated with faulty proteostasis, that promote the formation of protein aggregates, amyloid fibrils, intracellular, and extracellular inclusions in the most common nondegenerative disorders also referred to as protein misfolding disorders.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy.
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Pan- and isoform-specific inhibition of Hsp90: Design strategy and recent advances. Eur J Med Chem 2022; 238:114516. [DOI: 10.1016/j.ejmech.2022.114516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022]
|
26
|
Licata NV, Cristofani R, Salomonsson S, Wilson KM, Kempthorne L, Vaizoglu D, D’Agostino VG, Pollini D, Loffredo R, Pancher M, Adami V, Bellosta P, Ratti A, Viero G, Quattrone A, Isaacs AM, Poletti A, Provenzani A. C9orf72 ALS/FTD dipeptide repeat protein levels are reduced by small molecules that inhibit PKA or enhance protein degradation. EMBO J 2022; 41:e105026. [PMID: 34791698 PMCID: PMC8724771 DOI: 10.15252/embj.2020105026] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 11/09/2022] Open
Abstract
Intronic GGGGCC (G4C2) hexanucleotide repeat expansion within the human C9orf72 gene represents the most common cause of familial forms of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) (C9ALS/FTD). Repeat-associated non-AUG (RAN) translation of repeat-containing C9orf72 RNA results in the production of neurotoxic dipeptide-repeat proteins (DPRs). Here, we developed a high-throughput drug screen for the identification of positive and negative modulators of DPR levels. We found that HSP90 inhibitor geldanamycin and aldosterone antagonist spironolactone reduced DPR levels by promoting protein degradation via the proteasome and autophagy pathways respectively. Surprisingly, cAMP-elevating compounds boosting protein kinase A (PKA) activity increased DPR levels. Inhibition of PKA activity, by both pharmacological and genetic approaches, reduced DPR levels in cells and rescued pathological phenotypes in a Drosophila model of C9ALS/FTD. Moreover, knockdown of PKA-catalytic subunits correlated with reduced translation efficiency of DPRs, while the PKA inhibitor H89 reduced endogenous DPR levels in C9ALS/FTD patient-derived iPSC motor neurons. Together, our results suggest new and druggable pathways modulating DPR levels in C9ALS/FTD.
Collapse
Affiliation(s)
- Nausicaa V Licata
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| | - Sally Salomonsson
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLUCL Queen Square Institute of NeurologyLondonUK
| | - Katherine M Wilson
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLUCL Queen Square Institute of NeurologyLondonUK
| | - Liam Kempthorne
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLUCL Queen Square Institute of NeurologyLondonUK
| | - Deniz Vaizoglu
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLUCL Queen Square Institute of NeurologyLondonUK
| | - Vito G D’Agostino
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Daniele Pollini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Rosa Loffredo
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Michael Pancher
- HTS Core Facility, Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Valentina Adami
- HTS Core Facility, Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
- Department of MedicineNYU at Grossman School of MedicineNYUSA
| | - Antonia Ratti
- Department of NeurologyStroke Unit and Laboratory of NeuroscienceIstituto Auxologico Italiano, IRCCSMilanItaly
- Dipartimento di Biotecnologie Mediche e Medicina TraslazionaleUniversità degli Studi di MilanoMilanItaly
| | | | - Alessandro Quattrone
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Adrian M Isaacs
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLUCL Queen Square Institute of NeurologyLondonUK
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanItaly
| | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| |
Collapse
|
27
|
Saha T, van Vliet AA, Cui C, Macias JJ, Kulkarni A, Pham LN, Lawler S, Spanholtz J, Georgoudaki AM, Duru AD, Goldman A. Boosting Natural Killer Cell Therapies in Glioblastoma Multiforme Using Supramolecular Cationic Inhibitors of Heat Shock Protein 90. Front Mol Biosci 2021; 8:754443. [PMID: 34926577 PMCID: PMC8673718 DOI: 10.3389/fmolb.2021.754443] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/30/2021] [Indexed: 01/09/2023] Open
Abstract
Allogeneic natural killer (aNK) cell adoptive therapy has the potential to dramatically impact clinical outcomes of glioblastoma multiforme (GBM). However, in order to exert therapeutic activity, NK cells require tumor expression of ligands for activating receptors, such as MHC Class I peptide A/B (MICA/B) and ULBPs. Here, we describe the use of a blood-brain barrier (BBB) permissive supramolecular cationic drug vehicle comprising an inhibitor of the chaperone heat shock protein 90 (Hsp90), which sustains a cytotoxic effect on GBM cells, boosts the expression of MICA/B and ULBPs on the residual population, and augments the activity of clinical-grade aNK cells (GTA002). First, we identify Hsp90 mRNA transcription and gain of function as significantly upregulated in GBM compared to other central nervous system tumors. Through a rational chemical design, we optimize a radicicol supramolecular prodrug containing cationic excipients, SCI-101, which displays >2-fold increase in relative BBB penetration compared to less cationic formulations in organoids, in vitro. Using 2D and 3D biological models, we confirm SCI-101 sustains GBM cytotoxicity 72 h after drug removal and induces cell surface MICA/B protein and ULBP mRNA up to 200% in residual tumor cells compared to the naked drug alone without augmenting the shedding of MICA/B, in vitro. Finally, we generate and test the sequential administration of SCI-101 with a clinical aNK cell therapy, GTA002, differentiated and expanded from healthy umbilical cord blood CD34+ hematopoietic stem cells. Using a longitudinal in vitro model, we demonstrate >350% relative cell killing is achieved in SCI-101-treated cell lines compared to vehicle controls. In summary, these data provide a first-of-its-kind BBB-penetrating, long-acting inhibitor of Hsp90 with monotherapy efficacy, which improves response to aNK cells and thus may rapidly alter the treatment paradigm for patients with GBM.
Collapse
Affiliation(s)
- Tanmoy Saha
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | | | - Chunxiao Cui
- Xsphera Biosciences Inc., Boston, MA, United States
| | - Jorge Jimenez Macias
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Arpita Kulkarni
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Luu Nhat Pham
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Sean Lawler
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, United States
| | | | | | | | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States.,Glycostem Therapeutics B.V., Oss, Netherlands.,Cancer Immunology, Dana Farber/Harvard Cancer Center, Boston, MA, United States
| |
Collapse
|
28
|
Charif SE, Vassallu MF, Salvañal L, Igaz LM. Protein synthesis modulation as a therapeutic approach for amyotrophic lateral sclerosis and frontotemporal dementia. Neural Regen Res 2021; 17:1423-1430. [PMID: 34916412 PMCID: PMC8771112 DOI: 10.4103/1673-5374.330593] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Protein synthesis is essential for cells to perform life metabolic processes. Pathological alterations of protein content can lead to particular diseases. Cells have an intrinsic array of mechanisms and pathways that are activated when protein misfolding, accumulation, aggregation or mislocalization occur. Some of them (like the unfolded protein response) represent complex interactions between endoplasmic reticulum sensors and elongation factors that tend to increase expression of chaperone proteins and/or repress translation in order to restore protein homeostasis (also known as proteostasis). This is even more important in neurons, as they are very susceptible to harmful effects associated with protein overload and proteostatic mechanisms are less effective with age. Several neurodegenerative pathologies such as Alzheimer's, Parkinson's, and Huntington's diseases, amyotrophic lateral sclerosis and frontotemporal dementia exhibit a particular molecular signature of distinct, unbalanced protein overload. In amyotrophic lateral sclerosis and frontotemporal dementia, the majority of cases present intracellular inclusions of ubiquitinated transactive response DNA-binding protein of 43 kDa (TDP-43). TDP-43 is an RNA binding protein that participates in RNA metabolism, among other functions. Dysregulation of TDP-43 (e.g. aggregation and mislocalization) can dramatically affect neurons, and this has been linked to disease development. Expression of amyotrophic lateral sclerosis/frontotemporal dementia TDP-43-related mutations in cellular and animal models has been shown to recapitulate key features of the amyotrophic lateral sclerosis/frontotemporal dementia disease spectrum. These variants can be causative of degeneration onset and progression. Most neurodegenerative diseases (including amyotrophic lateral sclerosis and frontotemporal dementia) have no cure at the moment; however, modulating translation has recently emerged as an attractive approach that can be performed at several steps (i.e. regulating activation of initiation and elongation factors, inhibiting unfolded protein response activation or inducing chaperone expression and activity). This review focuses on the features of protein imbalance in neurodegenerative disorders and the relevance of developing therapeutical compounds aiming at restoring proteostasis. We strive to highlight the importance of research on drugs that, not only restore protein imbalance without compromising translational activity of cells, but are also as safe as possible for the patients.
Collapse
Affiliation(s)
- Santiago E Charif
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires -CONICET, Buenos Aires, Argentina
| | - M Florencia Vassallu
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires -CONICET, Buenos Aires, Argentina
| | - Lara Salvañal
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires -CONICET, Buenos Aires, Argentina
| | - Lionel M Igaz
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires -CONICET, Buenos Aires, Argentina
| |
Collapse
|
29
|
Simon C, Soga T, Okano HJ, Parhar I. α-Synuclein-mediated neurodegeneration in Dementia with Lewy bodies: the pathobiology of a paradox. Cell Biosci 2021; 11:196. [PMID: 34798911 PMCID: PMC8605528 DOI: 10.1186/s13578-021-00709-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is epitomized by the pathognomonic manifestation of α-synuclein-laden Lewy bodies within selectively vulnerable neurons in the brain. By virtue of prion-like inheritance, the α-synuclein protein inexorably undergoes extensive conformational metamorphoses and culminate in the form of fibrillar polymorphs, instigating calamitous damage to the brain's neuropsychological networks. This epiphenomenon is nebulous, however, by lingering uncertainty over the quasi "pathogenic" behavior of α-synuclein conformers in DLB pathobiology. Despite numerous attempts, a monolithic "α-synuclein" paradigm that is able to untangle the enigma enshrouding the clinicopathological spectrum of DLB has failed to emanate. In this article, we review conceptual frameworks of α-synuclein dependent cell-autonomous and non-autonomous mechanisms that are likely to facilitate the transneuronal spread of degeneration through the neuraxis. In particular, we describe how the progressive demise of susceptible neurons may evolve from cellular derangements perpetrated by α-synuclein misfolding and aggregation. Where pertinent, we show how these bona fide mechanisms may mutually accentuate α-synuclein-mediated neurodegeneration in the DLB brain.
Collapse
Affiliation(s)
- Christopher Simon
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Tomoko Soga
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
30
|
Chen KS, Menezes K, Rodgers JB, O’Hara DM, Tran N, Fujisawa K, Ishikura S, Khodaei S, Chau H, Cranston A, Kapadia M, Pawar G, Ping S, Krizus A, Lacoste A, Spangler S, Visanji NP, Marras C, Majbour NK, El-Agnaf OMA, Lozano AM, Culotti J, Suo S, Ryu WS, Kalia SK, Kalia LV. Small molecule inhibitors of α-synuclein oligomers identified by targeting early dopamine-mediated motor impairment in C. elegans. Mol Neurodegener 2021; 16:77. [PMID: 34772429 PMCID: PMC8588601 DOI: 10.1186/s13024-021-00497-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Parkinson's disease is a disabling neurodegenerative movement disorder characterized by dopaminergic neuron loss induced by α-synuclein oligomers. There is an urgent need for disease-modifying therapies for Parkinson's disease, but drug discovery is challenged by lack of in vivo models that recapitulate early stages of neurodegeneration. Invertebrate organisms, such as the nematode worm Caenorhabditis elegans, provide in vivo models of human disease processes that can be instrumental for initial pharmacological studies. METHODS To identify early motor impairment of animals expressing α-synuclein in dopaminergic neurons, we first used a custom-built tracking microscope that captures locomotion of single C. elegans with high spatial and temporal resolution. Next, we devised a method for semi-automated and blinded quantification of motor impairment for a population of simultaneously recorded animals with multi-worm tracking and custom image processing. We then used genetic and pharmacological methods to define the features of early motor dysfunction of α-synuclein-expressing C. elegans. Finally, we applied the C. elegans model to a drug repurposing screen by combining it with an artificial intelligence platform and cell culture system to identify small molecules that inhibit α-synuclein oligomers. Screen hits were validated using in vitro and in vivo mammalian models. RESULTS We found a previously undescribed motor phenotype in transgenic α-synuclein C. elegans that correlates with mutant or wild-type α-synuclein protein levels and results from dopaminergic neuron dysfunction, but precedes neuronal loss. Together with artificial intelligence-driven in silico and in vitro screening, this C. elegans model identified five compounds that reduced motor dysfunction induced by α-synuclein. Three of these compounds also decreased α-synuclein oligomers in mammalian neurons, including rifabutin which has not been previously investigated for Parkinson's disease. We found that treatment with rifabutin reduced nigrostriatal dopaminergic neurodegeneration due to α-synuclein in a rat model. CONCLUSIONS We identified a C. elegans locomotor abnormality due to dopaminergic neuron dysfunction that models early α-synuclein-mediated neurodegeneration. Our innovative approach applying this in vivo model to a multi-step drug repurposing screen, with artificial intelligence-driven in silico and in vitro methods, resulted in the discovery of at least one drug that may be repurposed as a disease-modifying therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Kevin S. Chen
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Krystal Menezes
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | | | - Darren M. O’Hara
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Nhat Tran
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Kazuko Fujisawa
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Seiya Ishikura
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Shahin Khodaei
- Donnelly Centre, University of Toronto, Toronto, ON Canada
| | - Hien Chau
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Anna Cranston
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Minesh Kapadia
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Grishma Pawar
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Susan Ping
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Aldis Krizus
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | | | | | - Naomi P. Visanji
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Connie Marras
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, Toronto, ON Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON Canada
| | - Nour K. Majbour
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Omar M. A. El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Andres M. Lozano
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON Canada
| | - Joseph Culotti
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
| | - Satoshi Suo
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - William S. Ryu
- Donnelly Centre, University of Toronto, Toronto, ON Canada
- Department of Physics, University of Toronto, Toronto, ON Canada
| | - Suneil K. Kalia
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON Canada
- KITE and CRANIA, University Health Network, Toronto, ON Canada
| | - Lorraine V. Kalia
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, ON Canada
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, Toronto, ON Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON Canada
| |
Collapse
|
31
|
Vermeulen K, Cools R, Briard E, Auberson Y, Schoepfer J, Koole M, Cawthorne C, Bormans G. Preclinical Evaluation of [ 11C]YC-72-AB85 for In Vivo Visualization of Heat Shock Protein 90 in Brain and Cancer with Positron Emission Tomography. ACS Chem Neurosci 2021; 12:3915-3927. [PMID: 34597516 DOI: 10.1021/acschemneuro.1c00508] [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] [Indexed: 11/29/2022] Open
Abstract
Aberrant Hsp90 has been implied in cancer and neurodegenerative disorders. The development of a suitable Hsp90 Positron emission tomography (PET) probe can provide in vivo quantification of the expression levels of Hsp90 as a biomarker for diagnosis and follow-up of cancer and central nervous system (CNS) disease progression. In this respect, [11C]YC-72-AB85 was evaluated as an Hsp90 PET probe in B16.F10 melanoma bearing mice and its brain uptake was determined in rats and nonhuman primate. In vitro binding of [11C]YC-72-AB85 to tissue slices of mouse B16.F10 melanoma, PC3 prostate carcinoma, and rodent brain was evaluated using autoradiography. Biodistribution of [11C]YC-72-AB85 was evaluated in healthy and B16.F10 melanoma mice. In vivo brain uptake was assessed by μPET studies in rats and a rhesus monkey. In vitro binding was deemed Hsp90-specific by blocking studies with heterologous Hsp90 inhibitors onalespib and SNX-0723. Saturable Hsp90 binding was observed in brain, tumor, blood, and blood-rich organs in mice. In combined pretreatment and displacement studies, reversible and Hsp90-specific binding of [11C]YC-72-AB85 was observed in rat brain. Dynamic μPET brain scans in baseline and blocking conditions in a rhesus monkey indicated Hsp90-specific binding. [11C]YC-72-AB85 is a promising PET tracer for in vivo visualization of Hsp90 in tumor and brain. Clear differences of Hsp90 binding to blood and blood-rich organs were observed in tumor vs control mice. Further, we clearly demonstrate, for the first time, binding to a saturable Hsp90 pool in brain of rats and a rhesus monkey.
Collapse
Affiliation(s)
- Koen Vermeulen
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- Radiobiology Unit & NURA, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Romy Cools
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Emmanuelle Briard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Yves Auberson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Joseph Schoepfer
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Michel Koole
- Nuclear Medicine & Molecular Imaging & MoSAIC, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine & Molecular Imaging & MoSAIC, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
32
|
Molecular chaperones and Parkinson's disease. Neurobiol Dis 2021; 160:105527. [PMID: 34626793 DOI: 10.1016/j.nbd.2021.105527] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive death of dopaminergic neurons in the substantia nigra and the formation of Lewy bodies (LBs). Mutations in PD-related genes lead to neuronal pathogenesis through various mechanisms, with known examples including SNCA/α-synuclein (PAKR1), Parkin (PARK2), PINK1 (PARK6), DJ-1 (PARK7), and LRRK2 (PARK8). Molecular chaperones/co-chaperones are proteins that aid the folding of other proteins into a functionally active conformation. It has been demonstrated that chaperones/co-chaperones interact with PD-related proteins and regulate their function in PD. HSP70, HSP90 and small heat shock proteins can prevent neurodegeneration by regulating α-syn misfolding, oligomerization and aggregation. The function of chaperones is regulated by co-chaperones such as HSP110, HSP40, HOP, CHIP, and BAG family proteins. Parkin, PINK1 and DJ-1 are PD-related proteins which are associated with mitochondrial function. Molecular chaperones regulate mitochondrial function and protein homeostasis by interacting with these PD-related proteins. This review discusses critical molecular chaperones/co-chaperones and PD-related proteins which contribute to the pathogenesis of PD, hoping to provide new molecular targets for therapeutic interventions to thwart the disease progression instead of only bringing symptomatic relief. Moreover, appreciating the critical role of chaperones in PD can also help us screen efficient biomarkers to identify PD at an early stage.
Collapse
|
33
|
All Roads Lead to Rome: Different Molecular Players Converge to Common Toxic Pathways in Neurodegeneration. Cells 2021; 10:cells10092438. [PMID: 34572087 PMCID: PMC8468417 DOI: 10.3390/cells10092438] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple neurodegenerative diseases (NDDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD) are being suggested to have common cellular and molecular pathological mechanisms, characterized mainly by protein misfolding and aggregation. These large inclusions, most likely, represent an end stage of a molecular cascade; however, the soluble misfolded proteins, which take part in earlier steps of this cascade, are the more toxic players. These pathological proteins, which characterize each specific disease, lead to the selective vulnerability of different neurons, likely resulting from a combination of different intracellular mechanisms, including mitochondrial dysfunction, ER stress, proteasome inhibition, excitotoxicity, oxidative damage, defects in nucleocytoplasmic transport, defective axonal transport and neuroinflammation. Damage within these neurons is enhanced by damage from the nonneuronal cells, via inflammatory processes that accelerate the progression of these diseases. In this review, while acknowledging the hallmark proteins which characterize the most common NDDs; we place specific focus on the common overlapping mechanisms leading to disease pathology despite these different molecular players and discuss how this convergence may occur, with the ultimate hope that therapies effective in one disease may successfully translate to another.
Collapse
|
34
|
Kayed R, Dettmer U, Lesné SE. Soluble endogenous oligomeric α-synuclein species in neurodegenerative diseases: Expression, spreading, and cross-talk. JOURNAL OF PARKINSON'S DISEASE 2021; 10:791-818. [PMID: 32508330 PMCID: PMC7458533 DOI: 10.3233/jpd-201965] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is growing recognition in the field of neurodegenerative diseases that mixed proteinopathies are occurring at greater frequency than originally thought. This is particularly true for three amyloid proteins defining most of these neurological disorders, amyloid-beta (Aβ), tau, and alpha-synuclein (αSyn). The co-existence and often co-localization of aggregated forms of these proteins has led to the emergence of concepts positing molecular interactions and cross-seeding between Aβ, tau, and αSyn aggregates. Amongst this trio, αSyn has received particular attention in this context during recent years due to its ability to modulate Aβ and tau aggregation in vivo, to interact at a molecular level with Aβ and tau in vivo and to cross-seed tau in mice. Here we provide a comprehensive, critical, and accessible review about the expression, role and nature of endogenous soluble αSyn oligomers because of recent developments in the understanding of αSyn multimerization, misfolding, aggregation, cross-talk, spreading and cross-seeding in neurodegenerative disorders, including Parkinson's disease, dementia with Lewy bodies, multiple system atrophy, Alzheimer's disease, and Huntington's disease. We will also discuss our current understanding about the relative toxicity of endogenous αSyn oligomers in vivo and in vitro, and introduce potential opportunities to counter their deleterious effects.
Collapse
Affiliation(s)
- Rakez Kayed
- Departments of Neurology & Neuroscience & Cell Biology & Anatomy, University of Texas Medical Branch Galveston, Galveston, TX, USA,George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch Galveston, Galveston, TX, USA
| | - Ulf Dettmer
- Department of Neurology, Harvard Medical School, Boston, MA, USA,Ann Romney Center for Neurologic Diseases, Harvard Medical School, Boston, MA, USA
| | - Sylvain E. Lesné
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA,Institute of Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA,Correspondence to: Sylvain E. Lesné, PhD, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN 55414, USA. Tel.: +1 612 626 8341; E-mail: ; Website: https://lesnelab.org
| |
Collapse
|
35
|
Vikhareva EA, Chicheva MM, Evgen’ev MB, Funikov SY, Deikin AV, Bachurin SO, Ustyugov AA. Effect of Heat Shock Proteins on Life Span and Behavioral Functions of Animals with FUS Proteinopathy. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421010141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Expanding the role of proteasome homeostasis in Parkinson's disease: beyond protein breakdown. Cell Death Dis 2021; 12:154. [PMID: 33542205 PMCID: PMC7862491 DOI: 10.1038/s41419-021-03441-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
Proteasome is the principal hydrolytic machinery responsible for the great majority of protein degradation. The past three decades have testified prominent advances about proteasome involved in almost every aspect of biological processes. Nonetheless, inappropriate increase or decrease in proteasome function is regarded as a causative factor in several diseases. Proteasome abundance and proper assembly need to be precisely controlled. Indeed, various neurodegenerative diseases including Parkinson's disease (PD) share a common pathological feature, intracellular protein accumulation such as α-synuclein. Proteasome activation may effectively remove aggregates and prevent the neurodegeneration in PD, which provides a potential application for disease-modifying treatment. In this review, we build on the valuable discoveries related to different types of proteolysis by distinct forms of proteasome, and how its regulatory and catalytic particles promote protein elimination. Additionally, we summarize the emerging ideas on the proteasome homeostasis regulation by targeting transcriptional, translational, and post-translational levels. Given the imbalanced proteostasis in PD, the strategies for intensifying proteasomal degradation are advocated as a promising approach for PD clinical intervention.
Collapse
|
37
|
Uddin MA, Akhter MS, Kubra KT, Whitaker KE, Shipley SL, Smith LM, Barabutis N. Hsp90 inhibition protects the brain microvascular endothelium against oxidative stress. BRAIN DISORDERS 2021; 1. [PMID: 33569547 PMCID: PMC7869856 DOI: 10.1016/j.dscb.2020.100001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The brain endothelium is an integral element of the blood-brain barrier (BBB). Dysfunction of this formation due to increased generation of reactive oxygen species (ROS) progresses the establishment of neurological disorders including stroke and traumatic brain injury. Heat shock protein 90 inhibitors are anti-inflammatory agents, and their activities are mediated, at least in part, by P53. This is a tumor suppressor protein which regulates the opposing activities of Rac1 and RhoA in the cellular cytoskeleton. In the present study we investigated the role of Hsp90 inhibitors in the H2O2-induced brain endothelium breakdown, by employing human cerebral microvascular endothelial cells (hCMEC/D3). Our findings suggest that H2O2 downregulates P53 by enhancing the P53 suppressor mouse double minute 2 homolog (MDM2), as well as by increasing the apyrimidinic endonuclease 1/redox factor 1 (APE1/Ref1). The H2O2 – triggered violation of the brain endothelium barrier was reflected in measurements of transendothelial resistance, and the increased expression of the key cytoskeletal modulators cofilin and myosin light chain 2 (MLC2). Treatment of the hCMEC/D3 cells with Hsp90 inhibitors counteracted those events, and reduced the generation of the hydrogen peroxide – induced reactive oxygen species. Hence, our study suggests that Hsp90 inhibition supports the BBB integrity, and may represent a promising therapeutic approach for disorders associated with brain endothelium breakdown; including COVID-19.
Collapse
Affiliation(s)
- Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Kathryn E Whitaker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Summer L Shipley
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Landon M Smith
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| |
Collapse
|
38
|
Visanji NP, Madan P, Lacoste AMB, Buleje I, Han Y, Spangler S, Kalia LV, Hensley Alford S, Marras C. Using artificial intelligence to identify anti-hypertensives as possible disease modifying agents in Parkinson's disease. Pharmacoepidemiol Drug Saf 2020; 30:201-209. [PMID: 33219601 DOI: 10.1002/pds.5176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/25/2020] [Accepted: 10/21/2020] [Indexed: 11/10/2022]
Abstract
PURPOSE Drug repurposing is an effective means of increasing treatment options for diseases, however identifying candidate molecules for the indication of interest from the thousands of approved drugs is challenging. We have performed a computational analysis of published literature to rank existing drugs according to predicted ability to reduce alpha synuclein (aSyn) oligomerization and analyzed real-world data to investigate the association between exposure to highly ranked drugs and PD. METHODS Using IBM Watson for Drug Discoveryâ (WDD) we identified several antihypertensive drugs that may reduce aSyn oligomerization. Using IBM MarketScanâ Research Databases we constructed a cohort of individuals with incident hypertension. We conducted univariate and multivariate Cox proportional hazard analyses (HR) with exposure as a time-dependent covariate. Diuretics were used as the referent group. Age at hypertension diagnosis, sex, and several comorbidities were included in multivariate analyses. RESULTS Multivariate results revealed inverse associations for time to PD diagnosis with exposure to the combination of the combination of angiotensin receptor II blockers (ARBs) and dihydropyridine calcium channel blockers (DHP-CCB) (HR = 0.55, p < 0.01) and angiotensin converting enzyme inhibitors (ACEi) and diuretics (HR = 0.60, p-value <0.01). Increased risk was observed with exposure to alpha-blockers alone (HR = 1.81, p < 0.001) and the combination of alpha-blockers and CCB (HR = 3.17, p < 0.05). CONCLUSIONS We present evidence that a computational approach can efficiently identify leads for disease-modifying drugs. We have identified the combination of ARBs and DHP-CCBs as of particular interest in PD.
Collapse
Affiliation(s)
- Naomi P Visanji
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | | - Italo Buleje
- Foundational Innovation, Health Care and Life Sciences, IBM Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Yanyan Han
- IBM Almaden Research Center, San Jose, California, USA
| | | | - Lorraine V Kalia
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | | | - Connie Marras
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
39
|
Du XY, Xie XX, Liu RT. The Role of α-Synuclein Oligomers in Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21228645. [PMID: 33212758 PMCID: PMC7697105 DOI: 10.3390/ijms21228645] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
α-synuclein (α-syn) is a protein associated with the pathogenesis of Parkinson’s disease (PD), the second most common neurodegeneration disease with no effective treatment. However, how α-syn drives the pathology of PD remains elusive. Recent studies suggest that α-syn oligomers are the primary cause of neurotoxicity and play a critical role in PD. In this review, we discuss the process of α-syn oligomers formation and the current understanding of the structures of oligomers. We also describe seed and propagation effects of oligomeric forms of α-syn. Then, we summarize the mechanism by which α-syn oligomers exert neurotoxicity and promote neurodegeneration, including mitochondrial dysfunction, endoplasmic reticulum stress, proteostasis dysregulation, synaptic impairment, cell apoptosis and neuroinflammation. Finally, we investigate treatment regimens targeting α-syn oligomers at present. Further research is needed to understand the structure and toxicity mechanism of different types of oligomers, so as to provide theoretical basis for the treatment of PD.
Collapse
Affiliation(s)
- Xiao-yu Du
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing 100190, China; (X.-y.D.); (X.-x.X.)
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi-xiu Xie
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing 100190, China; (X.-y.D.); (X.-x.X.)
| | - Rui-tian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing 100190, China; (X.-y.D.); (X.-x.X.)
- Correspondence: ; Tel.: +86-10-82545017
| |
Collapse
|
40
|
Frey B, AlOkda A, Jackson MP, Riguet N, Duce JA, Lashuel HA. Monitoring alpha-synuclein oligomerization and aggregation using bimolecular fluorescence complementation assays: What you see is not always what you get. J Neurochem 2020; 157:872-888. [PMID: 32772367 PMCID: PMC8246987 DOI: 10.1111/jnc.15147] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022]
Abstract
Bimolecular fluorescence complementation (BiFC) was introduced a decade ago as a method to monitor alpha‐synuclein (α‐syn) oligomerization in intact cells. Since then, several α‐syn BiFC cellular assays and animal models have been developed based on the assumption that an increase in the fluorescent signal correlates with increased α‐syn oligomerization or aggregation. Despite the increasing use of these assays and models in mechanistic studies, target validation and drug screening, there have been no reports that (1) validate the extent to which the BiFC fluorescent signal correlates with α‐syn oligomerization at the biochemical level; (2) provide a structural characterization of the oligomers and aggregates formed by the BiFC. To address this knowledge gap, we first analysed the expression level and oligomerization properties of the individual constituents of α‐syn‐Venus, one of the most commonly used BiFC systems, in HEK‐293 & SH‐SY5Y cells from three different laboratories using multiple biochemical approaches and techniques. Next, we investigated the biochemical and aggregation properties of α‐syn upon co‐expression of both BiFC fragments. Our results show that (1) the C‐terminal‐Venus fused to α‐syn (α‐syn‐Vc) is present in much lower abundance than its counterpart with N‐terminal‐Venus fused to α‐syn (Vn‐α‐syn); (2) Vn‐α‐syn exhibits a high propensity to form oligomers and higher‐order aggregates; and (3) the expression of either or both fragments does not result in the formation of α‐syn fibrils or cellular inclusions. Furthermore, our results suggest that only a small fraction of Vn‐α‐syn is involved in the formation of the fluorescent BiFC complex and that some of the fluorescent signal may arise from the association or entrapment of α‐syn‐Vc in Vn‐α‐syn aggregates. The fact that the N‐terminal fragment exists predominantly in an aggregated state also indicates that one must exercise caution when using this system to investigate α‐syn oligomerization in cells or in vivo. Altogether, our results suggest that cellular and animal models of oligomerization, aggregation and cell‐to‐cell transmission based on the α‐syn BiFC systems should be thoroughly characterized at the biochemical level to ensure that they reproduce the process of interest and measure what they are intended to measure. ![]()
Collapse
Affiliation(s)
- Bryan Frey
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Abdelrahman AlOkda
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthew P Jackson
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Nathan Riguet
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - James A Duce
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
41
|
Sanchez J, Carter TR, Cohen MS, Blagg BSJ. Old and New Approaches to Target the Hsp90 Chaperone. Curr Cancer Drug Targets 2020; 20:253-270. [PMID: 31793427 DOI: 10.2174/1568009619666191202101330] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/30/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
The 90-kDa heat shock protein (Hsp90) is a molecular chaperone that ensures cellular proteostasis by maintaining the folding, stabilization, activation, and degradation of over 400 client proteins. Hsp90 is not only critical for routine protein maintenance in healthy cells, but also during states of cellular stress, such as cancer and neurodegenerative diseases. Due to its ability to affect phosphorylation of numerous client proteins, inhibition of Hsp90 has been an attractive anticancer approach since the early 1990's, when researchers identified a druggable target on the amino terminus of Hsp90 for a variety of cancers. Since then, 17 Hsp90 inhibitors that target the chaperone's Nterminal domain, have entered clinical trials. None, however, have been approved thus far by the FDA as a cancer monotherapy. In these trials, a major limitation observed with Hsp90 inhibition at the N-terminal domain was dose-limiting toxicities and relatively poor pharmacokinetic profiles. Despite this, preclinical and clinical research continues to show that Hsp90 inhibitors effectively target cancer cell death and decrease tumor progression supporting the rationale for the development of novel Hsp90 inhibitors. Here, we present an in-depth overview of the Hsp90 inhibitors used in clinical trials. Finally, we present current shifts in the field related to targeting the carboxy-terminal domain of Hsp90 as well as to the development of isoform-selective inhibitors as a means to bypass the pitfalls of current Hsp90 inhibitors and improve clinical trial outcomes.
Collapse
Affiliation(s)
- Jackee Sanchez
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Trever R Carter
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Mark S Cohen
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States.,Department of Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
42
|
Kiechle M, von Einem B, Höfs L, Voehringer P, Grozdanov V, Markx D, Parlato R, Wiesner D, Mayer B, Sakk O, Baumann B, Lukassen S, Liss B, Ekici AB, Ludolph AC, Walther P, Ferger B, McLean PJ, Falkenburger BH, Weishaupt JH, Danzer KM. In Vivo Protein Complementation Demonstrates Presynaptic α-Synuclein Oligomerization and Age-Dependent Accumulation of 8-16-mer Oligomer Species. Cell Rep 2020; 29:2862-2874.e9. [PMID: 31775051 DOI: 10.1016/j.celrep.2019.10.089] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 09/16/2019] [Accepted: 10/22/2019] [Indexed: 11/28/2022] Open
Abstract
Intracellular accumulation of α-synuclein (α-syn) and formation of Lewy bodies are neuropathological characteristics of Parkinson's disease (PD) and related α-synucleinopathies. Oligomerization and spreading of α-syn from neuron to neuron have been suggested as key events contributing to the progression of PD. To directly visualize and characterize α-syn oligomerization and spreading in vivo, we generated two independent conditional transgenic mouse models based on α-syn protein complementation assays using neuron-specifically expressed split Gaussia luciferase or split Venus yellow fluorescent protein (YFP). These transgenic mice allow direct assessment of the quantity and subcellular distribution of α-syn oligomers in vivo. Using these mouse models, we demonstrate an age-dependent accumulation of a specific subtype of α-syn oligomers. We provide in vivo evidence that, although α-syn is found throughout neurons, α-syn oligomerization takes place at the presynapse. Furthermore, our mouse models provide strong evidence for a transsynaptic cell-to-cell transfer of de novo generated α-syn oligomers in vivo.
Collapse
Affiliation(s)
| | | | - Lennart Höfs
- Department of Neurology, Dresden University Medical Center, Dresden, Germany
| | - Patrizia Voehringer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Daniel Markx
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Rosanna Parlato
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| | - Diana Wiesner
- Department of Neurology, Ulm University, Ulm, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Olena Sakk
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Soeren Lukassen
- Institute of Human Genetics, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Birgit Liss
- Institute of Applied Physiology, Ulm University, Ulm, Germany; New College, University of Oxford, Oxford OX1 3BN, UK
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Boris Ferger
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | |
Collapse
|
43
|
Savyon M, Engelender S. SUMOylation in α-Synuclein Homeostasis and Pathology. Front Aging Neurosci 2020; 12:167. [PMID: 32670048 PMCID: PMC7330056 DOI: 10.3389/fnagi.2020.00167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
The accumulation and aggregation of α-synuclein are central to Parkinson’s disease (PD), yet the molecular mechanisms responsible for these events are not fully understood. Post-translational modifications of α-synuclein regulate several of its properties, including degradation, interaction with proteins and membranes, aggregation and toxicity. SUMOylation is a post-translational modification involved in various nuclear and extranuclear processes, such as subcellular protein targeting, mitochondrial fission and synaptic plasticity. Protein SUMOylation increases in response to several stressful situations, from viral infections to trauma. In this framework, an increasing amount of evidence has implicated SUMOylation in several neurodegenerative diseases, including PD. This review will discuss recent findings in the role of SUMOylation as a regulator of α-synuclein accumulation, aggregation and toxicity, and its possible implication in neurodegeneration that underlies PD.
Collapse
Affiliation(s)
- Mor Savyon
- Department of Biochemistry, The B. Rappaport Faculty of Medicine and Institute of Medical Research, Technion - Israel Institute of Technology, Haifa, Israel
| | - Simone Engelender
- Department of Biochemistry, The B. Rappaport Faculty of Medicine and Institute of Medical Research, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
44
|
Joshi N, Raveendran A, Nagotu S. Chaperones and Proteostasis: Role in Parkinson's Disease. Diseases 2020; 8:diseases8020024. [PMID: 32580484 PMCID: PMC7349525 DOI: 10.3390/diseases8020024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Proper folding to attain a defined three-dimensional structure is a prerequisite for the functionality of a protein. Improper folding that eventually leads to formation of protein aggregates is a hallmark of several neurodegenerative disorders. Loss of protein homeostasis triggered by cellular stress conditions is a major contributing factor for the formation of these toxic aggregates. A conserved class of proteins called chaperones and co-chaperones is implicated in maintaining the cellular protein homeostasis. Expanding the body of evidence highlights the role of chaperones as central mediators in the formation, de-aggregation and degradation of the aggregates. Altered expression and function of chaperones is associated with many neurodegenerative diseases including Parkinson’s disease. Several studies indicate that chaperones are at the center of the cause and effect cycle of this disease. An overview of the various chaperones that are associated with homeostasis of Parkinson’s disease-related proteins and their role in pathogenicity will be discussed in this review.
Collapse
|
45
|
Bohush A, Niewiadomska G, Weis S, Filipek A. HSP90 and Its Novel Co-Chaperones, SGT1 and CHP-1, in Brain of Patients with Parkinson's Disease and Dementia with Lewy Bodies. JOURNAL OF PARKINSONS DISEASE 2020; 9:97-107. [PMID: 30741686 PMCID: PMC6398563 DOI: 10.3233/jpd-181443] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Parkinson’s disease (PD) is a neurodegenerative disorder characterized by the presence of inclusions known as Lewy bodies in some brain regions. Lewy bodies consist of α-synuclein and many other proteins including chaperones. Objective: To learn more about the role of chaperone complexes in PD and a related disorder, i.e., dementia with Lewy bodies (DLB), in this work we analyzed the expression of HSP90 and its two quite recently identified co-chaperones, SGT1 and CHP-1, in selected brain regions from patients suffering from these diseases. Methods: To fulfill the aim of our study we used human material and applied immunohistochemistry, Western blot analysis and real time/quantitative PCR (RT-qPCR). Results: We have found that HSP90 mRNA level is higher in the temporal cortex of PD and in frontal cortex of DLB brains, even though level of protein does not change significantly. The mRNA level of SGT1 is higher in the frontal and temporal cortex of PD and in substantia nigra of DLB brains while no significant changes in the level of protein were noticed. Similarly, the mRNA level of CHP-1 was found to be higher in the frontal and temporal cortex of PD and in all examined regions i.e. substantia nigra, frontal and temporal cortex of DLB brains. In the case of CHP-1 the protein level was found to be higher in frontal cortex of PD and in all examined areas of DLB patients. Conclusions: Our data indicate that the level of HSP90, SGT1 and CHP-1 is upregulated in the majority of cases of PD and DLB, which suggests that the examined proteins might be involved in these pathologies.
Collapse
Affiliation(s)
- Anastasiia Bohush
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Grażyna Niewiadomska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Serge Weis
- Division of Neuropathology, Neuromed Campus, Kepler University Hospital, Johannes Kepler University, Linz, Austria
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
46
|
Elmatboly AM, Sherif AM, Deeb DA, Benmelouka A, Bin-Jumah MN, Aleya L, Abdel-Daim MM. The impact of proteostasis dysfunction secondary to environmental and genetic causes on neurodegenerative diseases progression and potential therapeutic intervention. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11461-11483. [PMID: 32072427 DOI: 10.1007/s11356-020-07914-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/28/2020] [Indexed: 06/10/2023]
Abstract
Aggregation of particular proteins in the form of inclusion bodies or plaques followed by neuronal death is a hallmark of neurodegenerative proteopathies such as primary Parkinsonism, Alzheimer's disease, Lou Gehrig's disease, and Huntington's chorea. Complex polygenic and environmental factors implicated in these proteopathies. Accumulation of proteins in these disorders indicates a substantial disruption in protein homeostasis (proteostasis). Proteostasis or cellular proteome homeostasis is attained by the synchronization of a group of cellular mechanisms called the proteostasis network (PN), which is responsible for the stability of the proteome and achieves the equilibrium between synthesis, folding, and degradation of proteins. In this review, we will discuss the different types of PN and the impact of PN component dysfunction on the four major neurodegenerative diseases mentioned earlier. Graphical abstract.
Collapse
Affiliation(s)
| | - Ahmed M Sherif
- Faculty of Medicine, Zagazig University, El-Sharkia, Egypt
| | - Dalia A Deeb
- Faculty of Medicine, Zagazig University, El-Sharkia, Egypt
| | - Amira Benmelouka
- Faculty of Medicine, University of Algiers, Sidi M'Hamed, Algeria
| | - May N Bin-Jumah
- Biology Department, College Of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon Cedex, France
| | - Mohamed M Abdel-Daim
- Department of Zoology, Science College, King Saud University, Riyadh, 11451, Saudi Arabia.
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
47
|
Teil M, Arotcarena ML, Faggiani E, Laferriere F, Bezard E, Dehay B. Targeting α-synuclein for PD Therapeutics: A Pursuit on All Fronts. Biomolecules 2020; 10:biom10030391. [PMID: 32138193 PMCID: PMC7175302 DOI: 10.3390/biom10030391] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's Disease (PD) is characterized both by the loss of dopaminergic neurons in the substantia nigra and the presence of cytoplasmic inclusions called Lewy Bodies. These Lewy Bodies contain the aggregated α-synuclein (α-syn) protein, which has been shown to be able to propagate from cell to cell and throughout different regions in the brain. Due to its central role in the pathology and the lack of a curative treatment for PD, an increasing number of studies have aimed at targeting this protein for therapeutics. Here, we reviewed and discussed the many different approaches that have been studied to inhibit α-syn accumulation via direct and indirect targeting. These analyses have led to the generation of multiple clinical trials that are either completed or currently active. These clinical trials and the current preclinical studies must still face obstacles ahead, but give hope of finding a therapy for PD with time.
Collapse
Affiliation(s)
- Margaux Teil
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Marie-Laure Arotcarena
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Emilie Faggiani
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Florent Laferriere
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Benjamin Dehay
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
48
|
Sanderson JB, De S, Jiang H, Rovere M, Jin M, Zaccagnini L, Hays Watson A, De Boni L, Lagomarsino VN, Young-Pearse TL, Liu X, Pochapsky TC, Hyman BT, Dickson DW, Klenerman D, Selkoe DJ, Bartels T. Analysis of α-synuclein species enriched from cerebral cortex of humans with sporadic dementia with Lewy bodies. Brain Commun 2020; 2:fcaa010. [PMID: 32280944 PMCID: PMC7130446 DOI: 10.1093/braincomms/fcaa010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/23/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Since researchers identified α-synuclein as the principal component of Lewy bodies and Lewy neurites, studies have suggested that it plays a causative role in the pathogenesis of dementia with Lewy bodies and other ‘synucleinopathies’. While α-synuclein dyshomeostasis likely contributes to the neurodegeneration associated with the synucleinopathies, few direct biochemical analyses of α-synuclein from diseased human brain tissue currently exist. In this study, we analysed sequential protein extracts from a substantial number of patients with neuropathological diagnoses of dementia with Lewy bodies and corresponding controls, detecting a shift of cytosolic and membrane-bound physiological α-synuclein to highly aggregated forms. We then fractionated aqueous extracts (cytosol) from cerebral cortex using non-denaturing methods to search for soluble, disease-associated high molecular weight species potentially associated with toxicity. We applied these fractions and corresponding insoluble fractions containing Lewy-type aggregates to several reporter assays to determine their bioactivity and cytotoxicity. Ultimately, high molecular weight cytosolic fractions enhances phospholipid membrane permeability, while insoluble, Lewy-associated fractions induced morphological changes in the neurites of human stem cell-derived neurons. While the concentrations of soluble, high molecular weight α-synuclein were only slightly elevated in brains of dementia with Lewy bodies patients compared to healthy, age-matched controls, these observations suggest that a small subset of soluble α-synuclein aggregates in the brain may drive early pathogenic effects, while Lewy body-associated α-synuclein can drive neurotoxicity.
Collapse
Affiliation(s)
- John B Sanderson
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Suman De
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.,UK Dementia Research Institute, Department of Chemistry, University of Cambridge, Cambridge CB2 0AH, UK
| | - Haiyang Jiang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matteo Rovere
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ming Jin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ludovica Zaccagnini
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| | - Aurelia Hays Watson
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| | - Laura De Boni
- UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| | - Valentina N Lagomarsino
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xinyue Liu
- Department of Chemistry, Rosenstiel Institute for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Thomas C Pochapsky
- Department of Chemistry, Rosenstiel Institute for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Bradley T Hyman
- Massachusetts General Hospital, Harvard Medical School, Department of Neurology, Massachusetts Institute for Neurodegenerative Disease, Boston, MA 02129, USA
| | - Dennis W Dickson
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL 32224, USA
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.,UK Dementia Research Institute, Department of Chemistry, University of Cambridge, Cambridge CB2 0AH, UK
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tim Bartels
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,UK Dementia Research Institute, Department of Neurology, University College London, London WC1E 6BT, UK
| |
Collapse
|
49
|
Gupta A, Bansal A, Hashimoto-Torii K. HSP70 and HSP90 in neurodegenerative diseases. Neurosci Lett 2020; 716:134678. [PMID: 31816334 PMCID: PMC7336893 DOI: 10.1016/j.neulet.2019.134678] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/27/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Abstract
Molecular chaperones have a role to stabilize proteins or assist them in reaching their native fold. Heat shock proteins (HSPs) are a family of molecular chaperons that protect proteins from cellular stress during the assembly of protein complexes and also prevent the proteins from aggregation and disassembly. The immediate increase of HSPs is crucial for cellular adaptation to environmental changes and protection of other proteins from denaturation, thereby maintaining the cellular homeostasis and increasing the longevity of an organism. HSP70 and HSP90 are the most studied HSPs in this very large HSP family. Notably, HSP90 also stabilizes the disease-related proteins in neurodegenerative disorders. Therefore, small molecules that inhibit the HSP90 but also increase the HSP70 has been tested as potential drugs for neurodegenerative disorders.
Collapse
Affiliation(s)
- Abha Gupta
- University Institute of Biotechnology, Chandigarh University, Gharuan, 140413, India
| | - Ankush Bansal
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA.
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA; Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
50
|
Elia LP, Reisine T, Alijagic A, Finkbeiner S. Approaches to develop therapeutics to treat frontotemporal dementia. Neuropharmacology 2020; 166:107948. [PMID: 31962288 DOI: 10.1016/j.neuropharm.2020.107948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/16/2019] [Accepted: 01/07/2020] [Indexed: 12/13/2022]
Abstract
Frontotemporal degeneration (FTD) is a complex disease presenting as a spectrum of clinical disorders with progressive degeneration of frontal and temporal brain cortices and extensive neuroinflammation that result in personality and behavior changes, and eventually, death. There are currently no effective therapies for FTD. While 60-70% of FTD patients are sporadic cases, the other 30-40% are heritable (familial) cases linked to mutations in several known genes. We focus here on FTD caused by mutations in the GRN gene, which encodes a secreted protein, progranulin (PGRN), that has diverse roles in regulating cell survival, immune responses, and autophagy and lysosome function in the brain. FTD-linked mutations in GRN reduce brain PGRN levels that lead to autophagy and lysosome dysfunction, TDP43 accumulation, excessive microglial activation, astrogliosis, and neuron death through still poorly understood mechanisms. PGRN insufficiency has also been linked to Alzheimer's disease (AD), and so the development of therapeutics for GRN-linked FTD that restore PGRN levels and function may have broader application for other neurodegenerative diseases. This review focuses on a strategy to increase PGRN to functional, healthy levels in the brain by identifying novel genetic and chemical modulators of neuronal PGRN levels. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Lisa P Elia
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA.
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, CA, USA
| | - Amela Alijagic
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA; Departments of Neurology and Physiology, UCSF, San Francisco, CA, USA.
| |
Collapse
|