1
|
Yuan H, Chen Z, Wan L, Peng L, Long Z, He L, Wang C, Qiu R, Tang B, Jiang H. Pathogenicity analysis of three SCA14-associated missense mutations in PRKCG gene of Chinese patients with ataxia. Gene 2025; 958:149483. [PMID: 40221062 DOI: 10.1016/j.gene.2025.149483] [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: 01/28/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Spinocerebellar ataxia type 14 (SCA14) is an autosomal dominant disorder characterized by progressive cerebellar dysfunction and neurodegeneration. To date, it is rarely reported in China. SCA14 is caused by mutations in the PRKCG gene, which encodes protein kinase C gamma (PKCγ). Although nearly eighty distinct mutations of PRKCG gene have been identified, the pathological mechanisms of SCA14 remain unclear. In this study, we performed whole exome sequencing to screen causative genes in patients with unexplained progressive cerebellar ataxias, and identified three PRKCG mutations (c.302A > G, p.H101R, c.520C > G, p.H174D and c.2063C > G, p.P688R) that have not been previously reported in Chinese patients with SCA14. To explore the pathogenicity and function of these SCA14-associated PRKCG mutations, HEK293T and HeLa cells were transfected with the plasmids of empty vector, wild-type PRKCG and indicated PRKCG mutants. Protein stability, aggregation propensity, phosphorylation status, mitochondrial function and cytotoxicity were then measured. We found that H101R mutant PKCγ protein is unstable, prone to aggregate, exhibits reduced basal phosphorylation, and is resistant to agonist-mediated dephosphorylation. Also, H101R mutant PKCγ protein could result in increased apoptosis and reduced cell viability. These findings are similar to other pathogenic mutations. Additionally, cellular mitochondrial dysfunction was observed for the first time in cells expressing mutant PKCγ. Together, we identified three PRKCG mutations, expanding the mutation spectrum of PRKCG in China. The c.302A > G, p.H101R variant is pathogenic and mitochondrial dysfunction is suggested involved in the pathogenesis of SCA14.
Collapse
Affiliation(s)
- Hongyu Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, Hunan, China
| | - Linliu Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhe Long
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Lang He
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunrong Wang
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China; National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, Hunan, China; Furong Laboratory, Central South University, Changsha, Hunan 410008, China; Brain Research Center, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
2
|
Campomayor NB, Kim HJ, Kim M. Pro-Oxidative and Inflammatory Actions of Extracellular Hemoglobin and Heme: Molecular Events and Implications for Alzheimer's and Parkinson Disease. Biomol Ther (Seoul) 2025; 33:235-248. [PMID: 39962769 PMCID: PMC11893490 DOI: 10.4062/biomolther.2024.224] [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: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 03/01/2025] Open
Abstract
Hemoglobin (Hb) and heme, which are typically confined within red blood cells (RBCs), are essential for intravascular transport of gases and nutrients. However, these molecules acquire secondary functions upon exposure to the extracellular environment. Hb and heme generate reactive oxygen species (ROS), which are potent pro-inflammatory agents that contribute to oxidative stress and cellular damage. These events are relevant to neurodegenerative processes, where oxidative stress, irregular deposition of protein aggregates, and chronic inflammation are key pathological features. Extracellular Hb, heme, and oxidative stress derived from hemorrhagic events or RBC lysis may contribute to increased blood-brain barrier (BBB) permeability. These events allow Hb and heme to interact with neuroimmune cells and pathological protein aggregates, further amplifying pro-inflammatory signaling and the progression of Alzheimer's disease (AD) and Parkinson disease (PD). Chronic neuroinflammation, oxidative stress, and mitochondrial dysfunction lead to neuronal degeneration. Here, we sought to elucidate the pro-oxidative and inflammatory actions of extracellular Hb and heme, emphasizing their potential impact on AD and PD development.
Collapse
Affiliation(s)
- Nicole Bon Campomayor
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
3
|
Baldensperger T, Preissler M, Becker CFW. Non-enzymatic posttranslational protein modifications in protein aggregation and neurodegenerative diseases. RSC Chem Biol 2025; 6:129-149. [PMID: 39722676 PMCID: PMC11667106 DOI: 10.1039/d4cb00221k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Highly reactive metabolic intermediates and other small molecules frequently react with amino acid side chains, leading to non-enzymatic posttranslational modifications (nPTMs) of proteins. The abundance of these modifications increases under high metabolic activity or stress conditions and can dramatically impact protein structure and function. Although protein quality control mechanisms typically mitigate the effects of these impaired proteins, in long-lived and degradation-resistant proteins, nPTMs accumulate. In some cases, such as cataract development and diabetes, clear links between nPTMs, aging, and disease progression have been established. In neurodegenerative diseases such as Alzheimer's and Parkinson's disease, a key question is whether accumulation of nPTMs is a cause or consequence of protein aggregation. This review focuses on major nPTMs found on proteins with central roles in neurodegenerative diseases such as α-synuclein, β-amyloid, and tau. We summarize current knowledge on the formation of these modifications and discuss their potential impact on disease onset and progression. Additionally, we examine what is known to date about how nPTMs impair cellular detoxification, repair, and degradation systems. Finally, we critically discuss the available methodologies to systematically investigate nPTMs at the molecular level and outline suitable approaches to study their effects on protein aggregation. We aim to foster more research into the role of nPTMs in neurodegeneration by adapting methodologies that have proven successful in studying enzymatic posttranslational modifications. Specifically, we advocate for site-specific incorporation of these modifications into target proteins using advanced chemical and molecular biology techniques.
Collapse
Affiliation(s)
- Tim Baldensperger
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
| | - Miriam Preissler
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem) Währinger Str. 42 1090 Vienna Austria
| | - Christian F W Becker
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
| |
Collapse
|
4
|
Uversky VN. How to drug a cloud? Targeting intrinsically disordered proteins. Pharmacol Rev 2024; 77:PHARMREV-AR-2023-001113. [PMID: 39433443 DOI: 10.1124/pharmrev.124.001113] [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: 06/16/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
Biologically active proteins/regions without stable structure (i.e., intrinsically disordered proteins and regions (IDPs and IDRs)) are commonly found in all proteomes. They have a unique functional repertoire that complements the functionalities of ordered proteins and domains. IDPs/IDRs are multifunctional promiscuous binders capable of folding at interaction with specific binding partners on a template- or context-dependent manner, many of which undergo liquid-liquid phase separation, leading to the formation of membrane-less organelles and biomolecular condensates. Many of them are frequently related to the pathogenesis of various human diseases. All this defines IDPs/IDRs as attractive targets for the development of novel drugs. However, their lack of unique structures, multifunctionality, binding promiscuity, and involvement in unusual modes of action preclude direct use of traditional structure-based drug design approaches for targeting IDPs/IDRs, and make disorder-based drug discovery for these "protein clouds" challenging. Despite all these complexities there is continuing progress in the design of small molecules affecting IDPs/IDRs. This article describes the major structural features of IDPs/IDRs and the peculiarities of the disorder-based functionality. It also discusses the roles of IDPs/IDRs in various pathologies, and shows why the approaches elaborated for finding drugs targeting ordered proteins cannot be directly used for the intrinsic disorder-based drug design, and introduces some novel methodologies suitable for these purposes. Finally, it emphasizes that regardless of their multifunctionality, binding promiscuity, lack of unique structures, and highly dynamic nature, "protein clouds" are principally druggable. Significance Statement Intrinsically disordered proteins and regions are highly abundant in nature, have multiple important biological functions, are commonly involved in the pathogenesis of a multitude of human diseases, and are therefore considered as very attractive drug targets. Although dealing with these unstructured multifunctional protein/regions is a challenging task, multiple innovative approaches have been designed to target them by small molecules.
Collapse
|
5
|
Mitchell AK, Bliss RR, Church FC. Exercise, Neuroprotective Exerkines, and Parkinson's Disease: A Narrative Review. Biomolecules 2024; 14:1241. [PMID: 39456173 PMCID: PMC11506540 DOI: 10.3390/biom14101241] [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: 08/09/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease in which treatment often includes an exercise regimen. Exercise is neuroprotective in animal models of PD, and, more recently, human clinical studies have verified exercise's disease-modifying effect. Aerobic exercise and resistance training improve many of PD's motor and non-motor symptoms, while neuromotor therapy and stretching/flexibility exercises positively contribute to the quality of life in people with PD. Therefore, understanding the role of exercise in managing this complex disorder is crucial. Exerkines are bioactive substances that are synthesized and released during exercise and have been implicated in several positive health outcomes, including neuroprotection. Exerkines protect neuronal cells in vitro and rodent PD models in vivo. Aerobic exercise and resistance training both increase exerkine levels in the blood, suggesting a role for exerkines in the neuroprotective theory. Many exerkines demonstrate the potential for protecting the brain against pathological missteps caused by PD. Every person (people) with Parkinson's (PwP) needs a comprehensive exercise plan tailored to their unique needs and abilities. Here, we provide an exercise template to help PwP understand the importance of exercise for treating PD, describe barriers confronting many PwP in their attempt to exercise, provide suggestions for overcoming these barriers, and explore the role of exerkines in managing PD. In conclusion, exercise and exerkines together create a powerful neuroprotective system that should contribute to slowing the chronic progression of PD.
Collapse
Affiliation(s)
- Alexandra K. Mitchell
- Department of Health Sciences, Division of Physical Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | | | - Frank C. Church
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
He M, Zhou X, Wang X. Glycosylation: mechanisms, biological functions and clinical implications. Signal Transduct Target Ther 2024; 9:194. [PMID: 39098853 PMCID: PMC11298558 DOI: 10.1038/s41392-024-01886-1] [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: 10/21/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Protein post-translational modification (PTM) is a covalent process that occurs in proteins during or after translation through the addition or removal of one or more functional groups, and has a profound effect on protein function. Glycosylation is one of the most common PTMs, in which polysaccharides are transferred to specific amino acid residues in proteins by glycosyltransferases. A growing body of evidence suggests that glycosylation is essential for the unfolding of various functional activities in organisms, such as playing a key role in the regulation of protein function, cell adhesion and immune escape. Aberrant glycosylation is also closely associated with the development of various diseases. Abnormal glycosylation patterns are closely linked to the emergence of various health conditions, including cancer, inflammation, autoimmune disorders, and several other diseases. However, the underlying composition and structure of the glycosylated residues have not been determined. It is imperative to fully understand the internal structure and differential expression of glycosylation, and to incorporate advanced detection technologies to keep the knowledge advancing. Investigations on the clinical applications of glycosylation focused on sensitive and promising biomarkers, development of more effective small molecule targeted drugs and emerging vaccines. These studies provide a new area for novel therapeutic strategies based on glycosylation.
Collapse
Affiliation(s)
- Mengyuan He
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
7
|
Wei W, Wu Q, Wang S, Dong C, Shao S, Zhang Z, Zhang X, Zhang X, Kan J, Liu F. Treatment with walnut peptide ameliorates memory impairment in zebrafish and rats: promoting the expression of neurotrophic factors and suppressing oxidative stress. Food Funct 2024; 15:8043-8052. [PMID: 38988249 DOI: 10.1039/d4fo00074a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Walnut peptide, a low molecular weight peptide separated from walnuts by enzymatic hydrolysis, is considered as a potential nutraceutical with a variety of biological activities. In this study, we characterized the walnut peptide prepared by alkaline protease hydrolysis and evaluated its neuroprotective effect in zebrafish and rat models of memory disorders. Series of concentrations of the walnut peptide were orally administered to zebrafish and rats to examine its impact on the behavior and biochemical indicators. The results showed that the oral administration of walnut peptide significantly ameliorated the behavioral performance in zebrafish exposed to bisphenol AF (1 μg mL-1) and rats exposed to alcohol (30% ethanol, 10 mL kg-1). Furthermore, the walnut peptide upregulated the expression of neurotrophic-related molecules in zebrafish, such as the brain-derived neurotrophic factor (BDNF) and the glial cell-derived neurotrophic factor (GDNF). In the rat brain, the walnut peptide increased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), while dramatically reduced malondialdehyde (MDA) level. Together, these findings elucidated that the walnut peptide might partially offset the declarative memory deficits via regulation of neurotrophic-related molecule expression and promotion of the antioxidant defense ability. Therefore, walnut peptide holds the potential for development into functional foods as a nutritional supplement for the management of certain neurodegenerative disorders.
Collapse
Affiliation(s)
- Wei Wei
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Qiming Wu
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, PR China.
| | - Shuai Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chuanmin Dong
- Institute of Scientific and Technical Information of Heze, Heze, Shandong, 274005, PR China
| | - Shujuan Shao
- Heze Administrative Examination and Approval Service Bureau, Heze, Shandong, 274000, PR China
| | - Zhao Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Xiping Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Xuejun Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Juntao Kan
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, PR China.
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
8
|
Bonsor M, Ammar O, Schnoegl S, Wanker EE, Silva Ramos E. Polyglutamine disease proteins: Commonalities and differences in interaction profiles and pathological effects. Proteomics 2024; 24:e2300114. [PMID: 38615323 DOI: 10.1002/pmic.202300114] [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: 11/30/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
Currently, nine polyglutamine (polyQ) expansion diseases are known. They include spinocerebellar ataxias (SCA1, 2, 3, 6, 7, 17), spinal and bulbar muscular atrophy (SBMA), dentatorubral-pallidoluysian atrophy (DRPLA), and Huntington's disease (HD). At the root of these neurodegenerative diseases are trinucleotide repeat mutations in coding regions of different genes, which lead to the production of proteins with elongated polyQ tracts. While the causative proteins differ in structure and molecular mass, the expanded polyQ domains drive pathogenesis in all these diseases. PolyQ tracts mediate the association of proteins leading to the formation of protein complexes involved in gene expression regulation, RNA processing, membrane trafficking, and signal transduction. In this review, we discuss commonalities and differences among the nine polyQ proteins focusing on their structure and function as well as the pathological features of the respective diseases. We present insights from AlphaFold-predicted structural models and discuss the biological roles of polyQ-containing proteins. Lastly, we explore reported protein-protein interaction networks to highlight shared protein interactions and their potential relevance in disease development.
Collapse
Affiliation(s)
- Megan Bonsor
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Orchid Ammar
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sigrid Schnoegl
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Erich E Wanker
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eduardo Silva Ramos
- Department of Neuroproteomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
9
|
Itkin T, Unger K, Barak Y, Yovel A, Stekolshchik L, Ego L, Aydinov Y, Gerchman Y, Sapir A. Exploiting the Unique Biology of Caenorhabditis elegans to Launch Neurodegeneration Studies in Space. ASTROBIOLOGY 2024; 24:579-589. [PMID: 38917419 DOI: 10.1089/ast.2023.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The 21st century is likely to be the first century in which large-scale short- and long-term space missions become common. Accordingly, an ever-increasing body of research is focusing on understanding the effects of current and future space expeditions on human physiology in health and disease. Yet the complex experimental environment, the small number of participants, and the high cost of space missions are among the primary factors that hinder a better understanding of the impact of space missions on human physiology. The goal of our research was to develop a cost-effective, compact, and easy-to-manipulate system to address questions related to human health and disease in space. This initiative was part of the Ramon SpaceLab program, an annual research-based learning program designed to cultivate high school students' involvement in space exploration by facilitating experiments aboard the International Space Station (ISS). In the present study, we used the nematode Caenorhabditis elegans (C. elegans), a well-suited model organism, to investigate the effect of space missions on neurodegeneration-related processes. Our study specifically focused on the level of aggregation of Huntington's disease-causing polyglutamine stretch-containing (PolyQ) proteins in C. elegans muscles, the canonical system for studying neurodegeneration in this organism. We compared animals expressing PolyQ proteins grown onboard the ISS with their genetically identical siblings grown on Earth and observed a significant difference in the number of aggregates between the two populations. Currently, it is challenging to determine whether this effect stems from developmental or morphological differences between the cultures or is a result of life in space. Nevertheless, our results serve as a proof of concept and open a new avenue for utilizing C. elegans to address various open questions in space studies, including the effects of space conditions on the onset and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tatyana Itkin
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Ksenia Unger
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Yair Barak
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Amit Yovel
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Liya Stekolshchik
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Linoy Ego
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Yana Aydinov
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Yoram Gerchman
- Department of Biology and the Environment, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Institute of Evolution, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Oranim Academic College, Kiryat Tivon, Israel
| | - Amir Sapir
- Department of Biology and the Environment, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
10
|
Henriques C, Lopes MM, Silva AC, Lobo DD, Badin RA, Hantraye P, Pereira de Almeida L, Nobre RJ. Viral-based animal models in polyglutamine disorders. Brain 2024; 147:1166-1189. [PMID: 38284949 DOI: 10.1093/brain/awae012] [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: 07/09/2023] [Revised: 11/26/2023] [Accepted: 12/30/2023] [Indexed: 01/30/2024] Open
Abstract
Polyglutamine disorders are a complex group of incurable neurodegenerative disorders caused by an abnormal expansion in the trinucleotide cytosine-adenine-guanine tract of the affected gene. To better understand these disorders, our dependence on animal models persists, primarily relying on transgenic models. In an effort to complement and deepen our knowledge, researchers have also developed animal models of polyglutamine disorders employing viral vectors. Viral vectors have been extensively used to deliver genes to the brain, not only for therapeutic purposes but also for the development of animal models, given their remarkable flexibility. In a time- and cost-effective manner, it is possible to use different transgenes, at varying doses, in diverse targeted tissues, at different ages, and in different species, to recreate polyglutamine pathology. This paper aims to showcase the utility of viral vectors in disease modelling, share essential considerations for developing animal models with viral vectors, and provide a comprehensive review of existing viral-based animal models for polyglutamine disorders.
Collapse
Affiliation(s)
- Carina Henriques
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Miguel M Lopes
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Ana C Silva
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Diana D Lobo
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Romina Aron Badin
- CEA, DRF, Institute of Biology François Jacob, Molecular Imaging Research Center (MIRCen), 92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, Université Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), 92265 Fontenay-aux-Roses, France
| | - Philippe Hantraye
- CEA, DRF, Institute of Biology François Jacob, Molecular Imaging Research Center (MIRCen), 92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, Université Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), 92265 Fontenay-aux-Roses, France
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Jorge Nobre
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
11
|
Stroganova I, Willenberg H, Tente T, Depraz Depland A, Bakels S, Rijs AM. Exploring the Aggregation Propensity of PHF6 Peptide Segments of the Tau Protein Using Ion Mobility Mass Spectrometry Techniques. Anal Chem 2024; 96:5115-5124. [PMID: 38517679 PMCID: PMC10993201 DOI: 10.1021/acs.analchem.3c04974] [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: 11/03/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/24/2024]
Abstract
Peptide and protein aggregation involves the formation of oligomeric species, but the complex interplay between oligomers of different conformations and sizes complicates their structural elucidation. Using ion mobility mass spectrometry (IM-MS), we aim to reveal these early steps of aggregation for the Ac-PHF6-NH2 peptide segment from tau protein, thereby distinguishing between different oligomeric species and gaining an understanding of the aggregation pathway. An important factor that is often neglected, but which can alter the aggregation propensity of peptides, is the terminal capping groups. Here, we demonstrate the use of IM-MS to probe the early stages of aggregate formation of Ac-PHF6-NH2, Ac-PHF6, PHF6-NH2, and uncapped PHF6 peptide segments. The aggregation propensity of the four PHF6 segments is confirmed using thioflavin T fluorescence assays and transmission electron microscopy. A novel approach based on post-IM fragmentation and quadrupole selection on the TIMS-Qq-ToF (trapped ion mobility) spectrometer was developed to enhance oligomer assignment, especially for the higher-order aggregates. This approach pushes the limits of IM identification of isobaric species, whose signatures appear closer to each other with increasing oligomer size, and provides new insights into the interpretation of IM-MS data. In addition, TIMS collision cross section values are compared with traveling wave ion mobility (TWIMS) data to evaluate potential instrumental bias in the trapped ion mobility results. The two IM-MS instrumental platforms are based on different ion mobility principles and have different configurations, thereby providing us with valuable insight into the preservation of weakly bound biomolecular complexes such as peptide aggregates.
Collapse
Affiliation(s)
- Iuliia Stroganova
- Division
of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical
Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam 1081 HV, The Netherlands
- Centre
for Analytical Sciences Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Hannah Willenberg
- Division
of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical
Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam 1081 HV, The Netherlands
| | - Thaleia Tente
- Division
of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical
Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam 1081 HV, The Netherlands
| | - Agathe Depraz Depland
- Division
of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical
Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam 1081 HV, The Netherlands
- Centre
for Analytical Sciences Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Sjors Bakels
- Division
of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical
Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam 1081 HV, The Netherlands
- Centre
for Analytical Sciences Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Anouk M. Rijs
- Division
of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical
Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam 1081 HV, The Netherlands
- Centre
for Analytical Sciences Amsterdam, Amsterdam 1098 XH, The Netherlands
| |
Collapse
|
12
|
Ansari NK, Rais A, Naeem A. Methotrexate for Drug Repurposing as an Anti-Aggregatory Agent to Mercuric Treated α-Chymotrypsinogen-A. Protein J 2024; 43:362-374. [PMID: 38431536 DOI: 10.1007/s10930-024-10187-z] [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] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Protein aggregation is related to numerous pathological conditions like Alzheimer's and Parkinson's disease. In our study, we have shown that an already existing FDA-approved drug; methotrexate (MTX) can be reprofiled on preformed α-chymotrypsinogen A (α-Cgn A) aggregates. The zymogen showed formation of aggregates upon interaction with mercuric ions, with increasing concentration of Hg2Cl2 (0-150 µM). The hike in ThT and ANS fluorescence concomitant with blue shift, bathochromic shift and the hyperchromic effect in the CR absorbance, RLS and turbidity measurements, substantiate the zymogen β-rich aggregate formation. The secondary structural alterations of α- Cgn A as analyzed by CD measurements, FTIR and Raman spectra showed the transformation of native β-barrel conformation to β-inter-molecular rich aggregates. The native α- Cgn A have about 30% α-helical content which was found to be about 3% in presence of mercuric ions suggesting the formation of aggregates. The amorphous aggregates were visualized by SEM. On incubation of Hg2Cl2 treated α- Cgn A with increasing concentration of the MTX resulted in reversing aggregates to the native-like structure. These results were supported by remarkable decrease in ThT and ANS fluorescence intensities and CR absorbance and also consistent with CD, FTIR, and Raman spectroscopy data. MTX was found to increase the α-helical content of the zymogen from 3 to 15% proposing that drug is efficient in disrupting the β-inter-molecular rich aggregates and reverting it to native like structure. The SEM images are in accordance with CD data showing the disintegration of aggregates. The most effective concentration of the drug was found to be 120 µM. Molecular docking analysis showed that MTX molecule was surrounded by the hydrophobic residues including Phe39, His40, Arg145, Tyr146, Thr151, Gly193, Ser195, and Gly216 and conventional hydrogen bonds, including Gln73 (bond length: 2.67Å), Gly142 (2.59Å), Thr144 (2.81Å), Asn150 (2.73Å), Asp153 (2.71Å), and Cys191 (2.53Å). This investigation will help to find the use of already existing drugs to cure protein misfolding-related abnormalities.
Collapse
Affiliation(s)
- Neha Kausar Ansari
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U.P, 202002, India
| | - Amaan Rais
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U.P, 202002, India
| | - Aabgeena Naeem
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, U.P, 202002, India.
| |
Collapse
|
13
|
Potapenko A, Davidson JM, Lee A, Laird AS. The deubiquitinase function of ataxin-3 and its role in the pathogenesis of Machado-Joseph disease and other diseases. Biochem J 2024; 481:461-480. [PMID: 38497605 PMCID: PMC11088879 DOI: 10.1042/bcj20240017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Machado-Joseph disease (MJD) is a devastating and incurable neurodegenerative disease characterised by progressive ataxia, difficulty speaking and swallowing. Consequently, affected individuals ultimately become wheelchair dependent, require constant care, and face a shortened life expectancy. The monogenic cause of MJD is expansion of a trinucleotide (CAG) repeat region within the ATXN3 gene, which results in polyglutamine (polyQ) expansion within the resultant ataxin-3 protein. While it is well established that the ataxin-3 protein functions as a deubiquitinating (DUB) enzyme and is therefore critically involved in proteostasis, several unanswered questions remain regarding the impact of polyQ expansion in ataxin-3 on its DUB function. Here we review the current literature surrounding ataxin-3's DUB function, its DUB targets, and what is known regarding the impact of polyQ expansion on ataxin-3's DUB function. We also consider the potential neuroprotective effects of ataxin-3's DUB function, and the intersection of ataxin-3's role as a DUB enzyme and regulator of gene transcription. Ataxin-3 is the principal pathogenic protein in MJD and also appears to be involved in cancer. As aberrant deubiquitination has been linked to both neurodegeneration and cancer, a comprehensive understanding of ataxin-3's DUB function is important for elucidating potential therapeutic targets in these complex conditions. In this review, we aim to consolidate knowledge of ataxin-3 as a DUB and unveil areas for future research to aid therapeutic targeting of ataxin-3's DUB function for the treatment of MJD and other diseases.
Collapse
Affiliation(s)
- Anastasiya Potapenko
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Jennilee M. Davidson
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Albert Lee
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Angela S. Laird
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
14
|
Sofińska K, Seweryn S, Skirlińska-Nosek K, Barbasz J, Lipiec E. Tip-enhanced Raman spectroscopy reveals the structural rearrangements of tau protein aggregates at the growth phase. NANOSCALE 2024; 16:5294-5301. [PMID: 38372161 DOI: 10.1039/d3nr06365h] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Tau protein aggregates inside neurons in the course of Alzheimer's disease (AD). Because of the enormous number of people suffering from AD, this disease has become one of the world's major health and social problems. The presence of tau lesions clearly correlates with cognitive impairments in AD patients, thus, tau is the target of potential treatments for AD, next to amyloid-β. The exact mechanism of tau aggregation has not been understood in detail so far; especially little is known about the structural rearrangements of tau aggregates at the growth phase. The research into tau conformation at each step of the aggregation pathway will contribute to the design of effective therapeutic approaches. To follow the secondary structure of individual tau aggregates at the growth phase, we applied tip-enhanced Raman spectroscopy (TERS). The nanospectroscopic approach enabled us to follow the structure of individual aggregates occurring in the subsequent phases of tau aggregation. We applied multivariate data analysis to extract the spectral differences for tau aggregates at different aggregation phases. Moreover, atomic force microscopy (AFM) allowed the tracking of the morphological alterations for species occurring with the progression of tau aggregation.
Collapse
Affiliation(s)
- Kamila Sofińska
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Krakow, Poland.
| | - Sara Seweryn
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Krakow, Poland.
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Krakow, Poland
| | - Katarzyna Skirlińska-Nosek
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Krakow, Poland.
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Krakow, Poland
| | - Jakub Barbasz
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland
| | - Ewelina Lipiec
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Krakow, Poland.
| |
Collapse
|
15
|
Siraj S, Yameen D, Bhati S, Athar T, Khan S, Bhattacharya J, Islam A, Haque MM. Sugar osmolyte inhibits and attenuates the fibrillogenesis in RNase A: An in vitro and in silico characterizations. Int J Biol Macromol 2023; 253:127378. [PMID: 37839601 DOI: 10.1016/j.ijbiomac.2023.127378] [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: 06/27/2023] [Revised: 09/07/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Mechanisms of protein aggregation are of immense interest in therapeutic biology and neurodegenerative medicine. Biochemical processes within the living cell occur in a highly crowded environment. The phenomenon of macromolecular crowding affects the diffusional and conformational dynamics of proteins and modulates their folding. Macromolecular crowding is reported to cause protein aggregation in some cases, so it is a cause of concern as it leads to a plethora of neurodegenerative disorders and systemic amyloidosis. To divulge the mechanism of aggregation, it is imperative to study aggregation in well-characterized model proteins in the presence of macromolecular crowder. One such protein is ribonuclease A (RNase A), which deciphers neurotoxic function in humans; therefore we decided to explore the amyloid fibrillogenesis of this thermodynamically stable protein. To elucidate the impact of crowder, dextran-70 and its monomer glucose on the aggregation profile of RNase-A various techniques such as Absorbance, Fluorescence, Fourier Transforms Infrared, Dynamic Light Scattering and circular Dichroism spectroscopies along with imaging techniques like Atomic Force Microscopy and Transmission Electron Microscopy were employed. Thermal aggregation and fibrillation were further promoted by dextran-70 while glucose counteracted the effect of the crowding agent in a concentration-dependent manner. This study shows that glucose provides stability to the protein and prevents fibrillation. Intending to combat aggregation, which is the hallmark of numerous late-onset neurological disorders and systemic amyloidosis, this investigation unveils that naturally occurring osmolytes or other co-solutes can be further exploited in novel drug design strategies.
Collapse
Affiliation(s)
- Seerat Siraj
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Daraksha Yameen
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Shivani Bhati
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Teeba Athar
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Salman Khan
- Translational Research Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | | | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Mohammad Mahfuzul Haque
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
16
|
Ferreira PA. Nucleocytoplasmic transport at the crossroads of proteostasis, neurodegeneration and neuroprotection. FEBS Lett 2023; 597:2567-2589. [PMID: 37597509 DOI: 10.1002/1873-3468.14722] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023]
Abstract
Nucleocytoplasmic transport comprises the multistep assembly, transport, and disassembly of protein and RNA cargoes entering and exiting nuclear pores. Accruing evidence supports that impairments to nucleocytoplasmic transport are a hallmark of neurodegenerative diseases. These impairments cause dysregulations in nucleocytoplasmic partitioning and proteostasis of nuclear transport receptors and client substrates that promote intracellular deposits - another hallmark of neurodegeneration. Disturbances in liquid-liquid phase separation (LLPS) between dense and dilute phases of biomolecules implicated in nucleocytoplasmic transport promote micrometer-scale coacervates, leading to proteinaceous aggregates. This Review provides historical and emerging principles of LLPS at the interface of nucleocytoplasmic transport, proteostasis, aging and noxious insults, whose dysregulations promote intracellular aggregates. E3 SUMO-protein ligase Ranbp2 constitutes the cytoplasmic filaments of nuclear pores, where it acts as a molecular hub for rate-limiting steps of nucleocytoplasmic transport. A vignette is provided on the roles of Ranbp2 in nucleocytoplasmic transport and at the intersection of proteostasis in the survival of photoreceptor and motor neurons under homeostatic and pathophysiological environments. Current unmet clinical needs are highlighted, including therapeutics aiming to manipulate aggregation-dissolution models of purported neurotoxicity in neurodegeneration.
Collapse
Affiliation(s)
- Paulo A Ferreira
- Department of Ophthalmology, Department of Pathology, Duke University Medical Center, NC, Durham, USA
| |
Collapse
|
17
|
Szała-Mendyk B, Phan TM, Mohanty P, Mittal J. Challenges in studying the liquid-to-solid phase transitions of proteins using computer simulations. Curr Opin Chem Biol 2023; 75:102333. [PMID: 37267850 PMCID: PMC10527940 DOI: 10.1016/j.cbpa.2023.102333] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 06/04/2023]
Abstract
"Membraneless organelles," also referred to as biomolecular condensates, perform a variety of cellular functions and their dysregulation is implicated in cancer and neurodegeneration. In the last two decades, liquid-liquid phase separation (LLPS) of intrinsically disordered and multidomain proteins has emerged as a plausible mechanism underlying the formation of various biomolecular condensates. Further, the occurrence of liquid-to-solid transitions within liquid-like condensates may give rise to amyloid structures, implying a biophysical link between phase separation and protein aggregation. Despite significant advances, uncovering the microscopic details of liquid-to-solid phase transitions using experiments remains a considerable challenge and presents an exciting opportunity for the development of computational models which provide valuable, complementary insights into the underlying phenomenon. In this review, we first highlight recent biophysical studies which provide new insights into the molecular mechanisms underlying liquid-to-solid (fibril) phase transitions of folded, disordered and multi-domain proteins. Next, we summarize the range of computational models used to study protein aggregation and phase separation. Finally, we discuss recent computational approaches which attempt to capture the underlying physics of liquid-to-solid transitions along with their merits and shortcomings.
Collapse
Affiliation(s)
- Beata Szała-Mendyk
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States.
| | - Tien Minh Phan
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States.
| | - Priyesh Mohanty
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States.
| | - Jeetain Mittal
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States; Department of Chemistry, Texas A&M University, TAMU 3255, College Station, 77843, Texas, United States; Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, TAMU 3255, College Station, 77843, Texas, United States.
| |
Collapse
|
18
|
Dendrimers in Neurodegenerative Diseases. Processes (Basel) 2023. [DOI: 10.3390/pr11020319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Parkinson’s Disease (PD), Alzheimer’s Disease (AD), Multiple Sclerosis (MS) and amyotrophic lateral sclerosis (ALS), are characterized by progressive loss of structure or function of neurons. Current therapies for NDs are only symptomatic and long-term ineffective. This challenge has promoted the development of new therapies against relevant targets in these pathologies. In this review, we will focus on the most promising therapeutic approaches based on dendrimers (DDs) specially designed for the treatment and diagnosis of NDs. DDs are well-defined polymeric structures that provide a multifunctional platform for developing different nanosystems for a myriad of applications. DDs have been proposed as interesting drug delivery systems with the ability to cross the blood–brain barrier (BBB) and increase the bioavailability of classical drugs in the brain, as well as genetic material, by reducing the synthesis of specific targets, as β-amyloid peptide. Moreover, DDs have been shown to be promising anti-amyloidogenic systems against amyloid-β peptide (Aβ) and Tau aggregation, powerful agents for blocking α-synuclein (α-syn) fibrillation, exhibit anti-inflammatory properties, promote cellular uptake to certain cell types, and are potential tools for ND diagnosis. In summary, DDs have emerged as promising alternatives to current ND therapies since they may limit the extent of damage and provide neuroprotection to the affected tissues.
Collapse
|
19
|
Monge FA, Fanni AM, Donabedian PL, Hulse J, Maphis NM, Jiang S, Donaldson TN, Clark BJ, Whitten DG, Bhaskar K, Chi EY. Selective In Vitro and Ex Vivo Staining of Brain Neurofibrillary Tangles and Amyloid Plaques by Novel Ethylene Ethynylene-Based Optical Sensors. BIOSENSORS 2023; 13:151. [PMID: 36831917 PMCID: PMC9953543 DOI: 10.3390/bios13020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
The identification of protein aggregates as biomarkers for neurodegeneration is an area of interest for disease diagnosis and treatment development. In this work, we present novel super luminescent conjugated polyelectrolyte molecules as ex vivo sensors for tau-paired helical filaments (PHFs) and amyloid-β (Aβ) plaques. We evaluated the use of two oligo-p-phenylene ethynylenes (OPEs), anionic OPE12- and cationic OPE24+, as stains for fibrillar protein pathology in brain sections of transgenic mouse (rTg4510) and rat (TgF344-AD) models of Alzheimer's disease (AD) tauopathy, and post-mortem brain sections from human frontotemporal dementia (FTD). OPE12- displayed selectivity for PHFs in fluorimetry assays and strong staining of neurofibrillary tangles (NFTs) in mouse and human brain tissue sections, while OPE24+ stained both NFTs and Aβ plaques. Both OPEs stained the brain sections with limited background or non-specific staining. This novel family of sensors outperformed the gold-standard dye Thioflavin T in sensing capacities and co-stained with conventional phosphorylated tau (AT180) and Aβ (4G8) antibodies. As the OPEs readily bind protein amyloids in vitro and ex vivo, they are selective and rapid tools for identifying proteopathic inclusions relevant to AD. Such OPEs can be useful in understanding pathogenesis and in creating in vivo diagnostically relevant detection tools for neurodegenerative diseases.
Collapse
Affiliation(s)
- Florencia A. Monge
- Biomedical Engineering Graduate Program, University of New Mexico, Albuquerque, NM 87131, USA
- Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
| | - Adeline M. Fanni
- Biomedical Engineering Graduate Program, University of New Mexico, Albuquerque, NM 87131, USA
- Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
| | - Patrick L. Donabedian
- Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
- Nanoscience and Microsystems Engineering Graduate Program, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jonathan Hulse
- Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Nicole M. Maphis
- Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Neuroscience, University of New Mexico, Albuquerque, NM 87131, USA
| | - Shanya Jiang
- Department of Neuroscience, University of New Mexico, Albuquerque, NM 87131, USA
- Sartorius, Bohemia, NY 11716, USA
| | - Tia N. Donaldson
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Benjamin J. Clark
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA
| | - David G. Whitten
- Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131, USA
| | - Kiran Bhaskar
- Department of Neuroscience, University of New Mexico, Albuquerque, NM 87131, USA
| | - Eva Y. Chi
- Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
20
|
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: 20] [Impact Index Per Article: 10.0] [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
|
21
|
Vignaroli F, Mele A, Tondo G, De Giorgis V, Manfredi M, Comi C, Mazzini L, De Marchi F. The Need for Biomarkers in the ALS-FTD Spectrum: A Clinical Point of View on the Role of Proteomics. Proteomes 2023; 11:proteomes11010001. [PMID: 36648959 PMCID: PMC9844364 DOI: 10.3390/proteomes11010001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are severely debilitating and progressive neurodegenerative disorders. A distinctive pathological feature of several neurodegenerative diseases, including ALS and FTD, is the deposition of aberrant protein inclusions in neuronal cells, which leads to cellular dysfunction and neuronal damage and loss. Despite this, to date, the biological process behind developing these protein inclusions must be better clarified, making the development of disease-modifying treatment impossible until this is done. Proteomics is a powerful tool to characterize the expression, structure, functions, interactions, and modifications of proteins of tissue and biological fluid, including plasma, serum, and cerebrospinal fluid. This protein-profiling characterization aims to identify disease-specific protein alteration or specific pathology-based mechanisms which may be used as markers of these conditions. Our narrative review aims to highlight the need for biomarkers and the potential use of proteomics in clinical practice for ALS-FTD spectrum disorders, considering the emerging rationale in proteomics for new drug development. Certainly, new data will emerge in the near future in this regard and support clinicians in the development of personalized medicine.
Collapse
Affiliation(s)
| | - Angelica Mele
- Neurology Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Giacomo Tondo
- Department of Neurology, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research and Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research and Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Cristoforo Comi
- Department of Neurology, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Letizia Mazzini
- Neurology Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Fabiola De Marchi
- Neurology Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Correspondence: ; Tel.: +39-0321-3733962
| |
Collapse
|
22
|
Kabir ER, Chowdhury NM, Yasmin H, Kabir MT, Akter R, Perveen A, Ashraf GM, Akter S, Rahman MH, Sweilam SH. Unveiling the Potential of Polyphenols as Anti-Amyloid Molecules in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:787-807. [PMID: 36221865 PMCID: PMC10227919 DOI: 10.2174/1570159x20666221010113812] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease that mostly affects the elderly population. Mechanisms underlying AD pathogenesis are yet to be fully revealed, but there are several hypotheses regarding AD. Even though free radicals and inflammation are likely to be linked with AD pathogenesis, still amyloid-beta (Aβ) cascade is the dominant hypothesis. According to the Aβ hypothesis, a progressive buildup of extracellular and intracellular Aβ aggregates has a significant contribution to the AD-linked neurodegeneration process. Since Aβ plays an important role in the etiology of AD, therefore Aβ-linked pathways are mainly targeted in order to develop potential AD therapies. Accumulation of Aβ plaques in the brains of AD individuals is an important hallmark of AD. These plaques are mainly composed of Aβ (a peptide of 39-42 amino acids) aggregates produced via the proteolytic cleavage of the amyloid precursor protein. Numerous studies have demonstrated that various polyphenols (PPHs), including cyanidins, anthocyanins, curcumin, catechins and their gallate esters were found to markedly suppress Aβ aggregation and prevent the formation of Aβ oligomers and toxicity, which is further suggesting that these PPHs might be regarded as effective therapeutic agents for the AD treatment. This review summarizes the roles of Aβ in AD pathogenesis, the Aβ aggregation pathway, types of PPHs, and distribution of PPHs in dietary sources. Furthermore, we have predominantly focused on the potential of food-derived PPHs as putative anti-amyloid drugs.
Collapse
Affiliation(s)
- Eva Rahman Kabir
- School of Pharmacy, BRAC University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | | | - Hasina Yasmin
- School of Pharmacy, BRAC University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Md. Tanvir Kabir
- School of Pharmacy, BRAC University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Dhaka, Bangladesh
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Shamima Akter
- Department of Bioinformatics and Computational Biology, George Mason University, Fairfax, Virginia 22030, USA
| | | | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City 11829, Egypt
| |
Collapse
|
23
|
Prodromou C, Aran-Guiu X, Oberoi J, Perna L, Chapple JP, van der Spuy J. HSP70-HSP90 Chaperone Networking in Protein-Misfolding Disease. Subcell Biochem 2023; 101:389-425. [PMID: 36520314 DOI: 10.1007/978-3-031-14740-1_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Molecular chaperones and their associated co-chaperones are essential in health and disease as they are key facilitators of protein-folding, quality control and function. In particular, the heat-shock protein (HSP) 70 and HSP90 molecular chaperone networks have been associated with neurodegenerative diseases caused by aberrant protein-folding. The pathogenesis of these disorders usually includes the formation of deposits of misfolded, aggregated protein. HSP70 and HSP90, plus their co-chaperones, have been recognised as potent modulators of misfolded protein toxicity, inclusion formation and cell survival in cellular and animal models of neurodegenerative disease. Moreover, these chaperone machines function not only in folding but also in proteasome-mediated degradation of neurodegenerative disease proteins. This chapter gives an overview of the HSP70 and HSP90 chaperones, and their respective regulatory co-chaperones, and explores how the HSP70 and HSP90 chaperone systems form a larger functional network and its relevance to counteracting neurodegenerative disease associated with misfolded proteins and disruption of proteostasis.
Collapse
Affiliation(s)
| | - Xavi Aran-Guiu
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Jasmeen Oberoi
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Laura Perna
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - J Paul Chapple
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | |
Collapse
|
24
|
Mahakud AK, Shaikh J, Rifa Iqbal VV, Gupta A, Tiwari A, Saleem M. Amyloids on Membrane Interfaces: Implications for Neurodegeneration. J Membr Biol 2022; 255:705-722. [PMID: 35670831 DOI: 10.1007/s00232-022-00245-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/12/2022] [Indexed: 12/24/2022]
Abstract
Membrane interfaces are vital for various cellular processes, and their involvement in neurodegenerative disorders such as Alzheimer's and Parkinson's disease has taken precedence in recent years. The amyloidogenic proteins associated with neurodegenerative diseases interact with the neuronal membrane through various means, which has implications for both the onset and progression of the disease. The parameters that regulate the interaction between the membrane and the amyloids remain poorly understood. The review focuses on the various aspects of membrane interactions of amyloids, particularly amyloid-β (Aβ) peptides and Tau involved in Alzheimer's and α-synuclein involved in Parkinson's disease. The genetic, cell biological, biochemical, and biophysical studies that form the basis for our current understanding of the membrane interactions of Aβ peptides, Tau, and α-synuclein are discussed.
Collapse
Affiliation(s)
- Amaresh Kumar Mahakud
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - Jafarulla Shaikh
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - V V Rifa Iqbal
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - Abhinav Gupta
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - Anuj Tiwari
- Department of Life Sciences, National Institute of Technology, Rourkela, India
| | - Mohammed Saleem
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India. .,Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
25
|
Wang L, Hsiung CH, Liu X, Wang S, Loredo A, Zhang X, Xiao H. Xanthone-based solvatochromic fluorophores for quantifying micropolarity of protein aggregates. Chem Sci 2022; 13:12540-12549. [PMID: 36382293 PMCID: PMC9629104 DOI: 10.1039/d2sc05004h] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/12/2022] [Indexed: 01/31/2023] Open
Abstract
Proper three-dimensional structures are essential for maintaining the functionality of proteins and for avoiding pathological consequences of improper folding. Misfolding and aggregation of proteins have been both associated with neurodegenerative disease. Therefore, a variety of fluorogenic tools that respond to both polarity and viscosity have been developed to detect protein aggregation. However, the rational design of highly sensitive fluorophores that respond solely to polarity has remained elusive. In this work, we demonstrate that electron-withdrawing heteroatoms with (d-p)-π* conjugation can stabilize lowest unoccupied molecular orbital (LUMO) energy levels and promote bathochromic shifts. Guided by computational analyses, we have devised a novel series of xanthone-based solvatochromic fluorophores that have rarely been systematically studied. The resulting probes exhibit superior sensitivity to polarity but are insensitive to viscosity. As proof of concept, we have synthesized protein targeting probes for live-cell confocal imaging intended to quantify the polarity of misfolded and aggregated proteins. Interestingly, our results reveal several layers of protein aggregates in a way that we had not anticipated. First, microenvironments with reduced polarity were validated in the misfolding and aggregation of folded globular proteins. Second, granular aggregates of AgHalo displayed a less polar environment than aggregates formed by folded globular protein represented by Htt-polyQ. Third, our studies reveal that granular protein aggregates formed in response to different types of stressors exhibit significant polarity differences. These results show that the solvatochromic fluorophores solely responsive to polarity represent a new class of indicators that can be widely used for detecting protein aggregation in live cells, thus paving the way for elucidating cellular mechanisms of protein aggregation as well as therapeutic approaches to managing intracellular aggregates.
Collapse
Affiliation(s)
- Lushun Wang
- Department of Chemistry, Rice University 6100 Main Street Houston Texas 77005 USA
| | - Chia-Heng Hsiung
- Department of Chemistry, Pennsylvania State University, University Park PA 16802 USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park PA 16802 USA
| | - Xiaojing Liu
- Department of Chemistry, Pennsylvania State University, University Park PA 16802 USA
| | - Shichao Wang
- Department of Chemistry, Rice University 6100 Main Street Houston Texas 77005 USA
| | - Axel Loredo
- Department of Chemistry, Rice University 6100 Main Street Houston Texas 77005 USA
| | - Xin Zhang
- Department of Chemistry, Pennsylvania State University, University Park PA 16802 USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park PA 16802 USA
| | - Han Xiao
- Department of Chemistry, Rice University 6100 Main Street Houston Texas 77005 USA
- Department of Biosciences, Rice University 6100 Main Street Houston Texas 77005 USA
- Department of Bioengineering, Rice University 6100 Main Street Houston Texas 77005 USA
| |
Collapse
|
26
|
Bae D, Jones RE, Piscopo KM, Tyagi M, Shepherd JD, Hollien J. Regulation of Blos1 by IRE1 prevents the accumulation of Huntingtin protein aggregates. Mol Biol Cell 2022; 33:ar125. [PMID: 36044348 PMCID: PMC9634971 DOI: 10.1091/mbc.e22-07-0281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Huntington's disease is characterized by accumulation of the aggregation-prone mutant Huntingtin (mHTT) protein. Here, we show that expression of exon 1 of mHTT in mouse cultured cells activates IRE1, the transmembrane sensor of stress in the endoplasmic reticulum, leading to degradation of the Blos1 mRNA and repositioning of lysosomes and late endosomes toward the microtubule organizing center. Overriding Blos1 degradation results in excessive accumulation of mHTT aggregates in both cultured cells and primary neurons. Although mHTT is degraded by macroautophagy when highly expressed, we show that before the formation of large aggregates, mHTT is degraded via an ESCRT-dependent, macroautophagy-independent pathway consistent with endosomal microautophagy. This pathway is enhanced by Blos1 degradation and appears to protect cells from a toxic, less aggregated form of mHTT.
Collapse
Affiliation(s)
- Donghwi Bae
- School of Biological Sciences and Center for Cell and Genome Science, and
| | | | | | - Mitali Tyagi
- Department of Neurobiology, School of Medicine, University of Utah, Salt Lake City, UT 84112
| | - Jason D. Shepherd
- Department of Neurobiology, School of Medicine, University of Utah, Salt Lake City, UT 84112
| | - Julie Hollien
- School of Biological Sciences and Center for Cell and Genome Science, and,*Address correspondence to: Julie Hollien ()
| |
Collapse
|
27
|
Jiao F, Zhou B, Meng L. The regulatory mechanism and therapeutic potential of transcription factor EB in neurodegenerative diseases. CNS Neurosci Ther 2022; 29:37-59. [PMID: 36184826 PMCID: PMC9804079 DOI: 10.1111/cns.13985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/29/2022] [Accepted: 09/14/2022] [Indexed: 02/06/2023] Open
Abstract
The autophagy-lysosomal pathway (ALP) is involved in the degradation of protein aggregates and damaged organelles. Transcription factor EB (TFEB), a major regulator of ALP, has emerged as a leading factor in addressing neurodegenerative disease pathology, including Alzheimer's disease (AD), Parkinson's disease (PD), PolyQ diseases, and Amyotrophic lateral sclerosis (ALS). In this review, we delineate the regulation of TFEB expression and its functions in ALP. Dysfunctions of TFEB and its role in the pathogenesis of several neurodegenerative diseases are reviewed. We summarize the protective effects and molecular mechanisms of some TFEB-targeted agonists in neurodegenerative diseases. We also offer our perspective on analyzing the pros and cons of these agonists in the treatment of neurodegenerative diseases from the perspective of drug development. More studies on the regulatory mechanisms of TFEB in other biological processes will aid our understanding of the application of TFEB-targeted therapy in neurodegeneration.
Collapse
Affiliation(s)
- Fengjuan Jiao
- School of Mental HealthJining Medical UniversityJiningChina,Shandong Key Laboratory of Behavioral Medicine, School of Mental HealthJining Medical UniversityJiningChina
| | - Bojie Zhou
- School of Mental HealthJining Medical UniversityJiningChina,Shandong Key Laboratory of Behavioral Medicine, School of Mental HealthJining Medical UniversityJiningChina
| | - Lingyan Meng
- School of Mental HealthJining Medical UniversityJiningChina,Shandong Key Laboratory of Behavioral Medicine, School of Mental HealthJining Medical UniversityJiningChina
| |
Collapse
|
28
|
Dehabadi MH, Caflisch A, Ilie IM, Firouzi R. Interactions of Curcumin's Degradation Products with the Aβ 42 Dimer: A Computational Study. J Phys Chem B 2022; 126:7627-7637. [PMID: 36148988 DOI: 10.1021/acs.jpcb.2c05846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Amyloid-β (Aβ) dimers are the smallest toxic species along the amyloid-aggregation pathway and among the most populated oligomeric accumulations present in the brain affected by Alzheimer's disease (AD). A proposed therapeutic strategy to avoid the aggregation of Aβ into higher-order structures is to develop molecules that inhibit the early stages of aggregation, i.e., dimerization. Under physiological conditions, the Aβ dimer is highly dynamic and does not attain a single well-defined structure but is rather characterized by an ensemble of conformations. In a recent study, a highly heterogeneous library of conformers of the Aβ dimer was generated by an efficient sampling method with constraints based on ion mobility mass spectrometry data. Here, we make use of the Aβ dimer library to study the interaction with two curcumin degradation products, ferulic aldehyde and vanillin, by molecular dynamics (MD) simulations. Ensemble docking and MD simulations are used to provide atomistic detail of the interactions between the curcumin degradation products and the Aβ dimer. The simulations show that the aromatic residues of Aβ, and in particular 19FF20, interact with ferulic aldehyde and vanillin through π-π stacking. The binding of these small molecules induces significant changes on the 16KLVFF20 region.
Collapse
Affiliation(s)
- Maryam Haji Dehabadi
- Department of Physical Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Pajohesh Boulevard, 1496813151 Tehran, Iran
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Ioana M Ilie
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Rohoullah Firouzi
- Department of Physical Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Pajohesh Boulevard, 1496813151 Tehran, Iran
| |
Collapse
|
29
|
Shi X, Wei T, Hu Y, Wang M, Tang Y. The associations between plasma soluble Trem1 and neurological diseases: a Mendelian randomization study. J Neuroinflammation 2022; 19:218. [PMID: 36068612 PMCID: PMC9446564 DOI: 10.1186/s12974-022-02582-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/27/2022] [Indexed: 12/08/2022] Open
Abstract
Background Triggering receptor expressed on myeloid cell 1 (Trem1) is an important regulator of cellular inflammatory responses. Neuroinflammation is a common thread across various neurological diseases. Soluble Trem1 (sTrem1) in plasma is associated with the development of central nervous system disorders. However, the extent of any causative effects of plasma sTrem1 on the risk of these disorders is still unclear. Method Genetic variants for plasma sTrem1 levels were selected as instrumental variables. Summary-level statistics of neurological disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), epilepsy, cerebrovascular diseases, and migraine were collected from genome-wide association studies (GWASs). Whether plasma sTrem1 was causally associated with neurological disorders was assessed using a two-sample Mendelian randomization (MR) analysis, with false discovery rate (FDR)-adjusted methods applied. Results We inferred suggestive association of higher plasma sTrem1 with the risk of AD (odds ratio [OR] per one standard deviation [SD] increase = 1.064, 95% CI 1.012–1.119, P = 0.014, PFDR = 0.056). Moreover, there was significant association between plasma sTrem1 level and the risk of epilepsy (OR per one SD increase = 1.044, 95% CI 1.016–1.072, P = 0.002, PFDR = 0.032), with a modest statistical power of 41%. Null associations were found for plasma sTrem1 with other neurological diseases and their subtypes. Conclusions Taken together, this study indicates suggestive association between plasma sTrem1 and AD. Moreover, higher plasma sTrem1 was associated with the increased risk of epilepsy. The findings support the hypothesis that sTrem1 may be a vital element on the causal pathway to AD and epilepsy. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02582-z.
Collapse
Affiliation(s)
- Xiaolei Shi
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Tao Wei
- Innovation Center for Neurological Disorders, Department of Neurology, National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yachun Hu
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.,Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meng Wang
- Innovation Center for Neurological Disorders, Department of Neurology, National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China. .,Neurodegenerative Laboratory of Ministry of Education of the Peoples Republic of China, Beijing, China.
| |
Collapse
|
30
|
4-Phenylbutyric Acid (4-PBA) Derivatives Prevent SOD1 Amyloid Aggregation In Vitro with No Effect on Disease Progression in SOD1-ALS Mice. Int J Mol Sci 2022; 23:ijms23169403. [PMID: 36012668 PMCID: PMC9409193 DOI: 10.3390/ijms23169403] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 12/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the degeneration of motor neurons. Mutations in the superoxide dismutase (SOD1) gene, causing protein misfolding and aggregation, were suggested as the pathogenic mechanisms involved in familial ALS cases. In the present study, we investigated the potential therapeutic effect of C4 and C5, two derivatives of the chemical chaperone 4-phenylbutyric acid (4-PBA). By combining in vivo and in vitro techniques, we show that, although C4 and C5 successfully inhibited amyloid aggregation of recombinant mutant SOD1 in a dose-dependent manner, they failed to suppress the accumulation of misfolded SOD1. Moreover, C4 or C5 daily injections to SOD1G93A mice following onset had no effect on either the accumulation of misfolded SOD1 or the neuroinflammatory response in the spinal cord and, consequently, failed to extend the survival of SOD1G93A mice or to improve their motor symptoms. Finally, pharmacokinetic (PK) studies demonstrated that high concentrations of C4 and C5 reached the brain and spinal cord but only for a short period of time. Thus, our findings suggest that use of such chemical chaperones for ALS drug development may need to be optimized for more effective results.
Collapse
|
31
|
Xu S, Li G, Ye X, Chen D, Chen Z, Xu Z, Daniele M, Tambone S, Ceccacci A, Tomei L, Ye L, Yu Y, Solbach A, Farmer SM, Stimming EF, McAllister G, Marchionini DM, Zhang S. HAP40 is a conserved central regulator of Huntingtin and a potential modulator of Huntington's disease pathogenesis. PLoS Genet 2022; 18:e1010302. [PMID: 35853002 PMCID: PMC9295956 DOI: 10.1371/journal.pgen.1010302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/18/2022] [Indexed: 11/19/2022] Open
Abstract
Perturbation of huntingtin (HTT)'s physiological function is one postulated pathogenic factor in Huntington's disease (HD). However, little is known how HTT is regulated in vivo. In a proteomic study, we isolated a novel ~40kDa protein as a strong binding partner of Drosophila HTT and demonstrated it was the functional ortholog of HAP40, an HTT associated protein shown recently to modulate HTT's conformation but with unclear physiological and pathologic roles. We showed that in both flies and human cells, HAP40 maintained conserved physical and functional interactions with HTT. Additionally, loss of HAP40 resulted in similar phenotypes as HTT knockout. More strikingly, HAP40 strongly affected HTT's stability, as depletion of HAP40 significantly reduced the levels of endogenous HTT protein while HAP40 overexpression markedly extended its half-life. Conversely, in the absence of HTT, the majority of HAP40 protein were degraded, likely through the proteasome. Further, the affinity between HTT and HAP40 was not significantly affected by polyglutamine expansion in HTT, and contrary to an early report, there were no abnormal accumulations of endogenous HAP40 protein in HD cells from mouse HD models or human patients. Lastly, when tested in Drosophila models of HD, HAP40 partially modulated the neurodegeneration induced by full-length mutant HTT while showed no apparent effect on the toxicity of mutant HTT exon 1 fragment. Together, our study uncovers a conserved mechanism governing the stability and in vivo functions of HTT and demonstrates that HAP40 is a central and positive regulator of endogenous HTT. Further, our results support that mutant HTT is toxic regardless of the presence of its partner HAP40, and implicate HAP40 as a potential modulator of HD pathogenesis through its multiplex effect on HTT's function, stability and the potency of mutant HTT's toxicity.
Collapse
Affiliation(s)
- Shiyu Xu
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Gang Li
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Xin Ye
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Dongsheng Chen
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Zhihua Chen
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Zhen Xu
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Moretti Daniele
- Department of Translational and Discovery Research, IRBM SpA, Pomezia (RM), Italy
| | - Sara Tambone
- Department of Translational and Discovery Research, IRBM SpA, Pomezia (RM), Italy
| | - Alessandra Ceccacci
- Department of Translational and Discovery Research, IRBM SpA, Pomezia (RM), Italy
| | - Licia Tomei
- Department of Translational and Discovery Research, IRBM SpA, Pomezia (RM), Italy
| | - Lili Ye
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - Yue Yu
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Programs in Genetics and Epigenetics and Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Amanda Solbach
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Programs in Genetics and Epigenetics and Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Stephen M. Farmer
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Program in Biochemistry and Cell Biology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
| | - Erin Furr Stimming
- Department of Neurology, HDSA Center of Excellence, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| | - George McAllister
- CHDI Management/CHDI Foundation, 350 Seventh Ave, New York, New York, United States of America
| | - Deanna M. Marchionini
- CHDI Management/CHDI Foundation, 350 Seventh Ave, New York, New York, United States of America
| | - Sheng Zhang
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
- Programs in Genetics and Epigenetics and Neuroscience, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (MD Anderson UTHealth GSBS), Houston, Texas, United States of America
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States of America
| |
Collapse
|
32
|
Mei J, Yang H, Ahmad S, Ma X, Xu W, Gao W, Li Y, Wang C, Ai H. Toxicity Mechanism of Aβ42 Oligomer in the Binding between the GABA BR1a sushi1 Domain and Amyloid Precursor Protein 9mer: A Mechanism like Substitution Reaction. ACS Chem Neurosci 2022; 13:2048-2059. [PMID: 35737468 DOI: 10.1021/acschemneuro.2c00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Amyloid-β peptide (Aβ), characterized by its abnormal folding into neurotoxic aggregates, impairs synaptic plasticity and causes synaptic loss associated with Alzheimer's disease (AD). The neurotoxicity of Aβ oligomers via the binding to various cell-surface receptors was frequently observed experimentally; however, the toxic mechanism still remains unknown. In this paper, we study the intervention of Aβ oligomers to the receptor-peptide binding in the GABABR1a sushi1-APP 9mer complex, a key node in increasing short-term synaptic facilitation in the mouse hippocampus and decreasing neuronal activity by inhibiting neurotransmitter release by molecular dynamics simulations. The residue types of Aβ42 oligomers involved in the intervention and core contact areas of the receptor were first identified, by which an unprecedented toxicity mechanism of Aβ42 oligomers is proposed. These involved residues of Aβ42 oligomers are positively charged residues Asp and Glu, and the core area of GABABR1a sushi1 domain is the Coil one, sharing the rich negatively charged residues R19/R21/R25/R45 with the pocket, in which APP 9mer is locked. The presence of an Aβ42 oligomer rather than of a monomer stretches these key residues in the core area and consequently "unlocks and releases" the APP 9mer from its initial pocket, unsteadying the sushi1 domain and taking into toxic effect. It looks like a chemical "substitution" reaction, Aβ42 oligomer + GABABR1a sushi1-APP 9mer complex → Aβ42 oligomer-GABABR1a sushi1 complex + APP 9mer. Further analysis reveals that the toxicity of Aβ42 oligomer to GABABR1a sushi1 domain stability depends on the residue number of the contact area and the size of Aβ42 oligomer, in which semi-extended trimeric Aβ42 oligomer is identified as the most toxic one. This work provides a novel insight into the mechanism of Aβ oligomeric toxicity to neuroreceptors and sets an important precedent for dealing with Aβ oligomeric toxicity to other receptors at the molecular level.
Collapse
Affiliation(s)
- Jinfei Mei
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Huijuan Yang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Sajjad Ahmad
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Xiaohong Ma
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Wen Xu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Wenqi Gao
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Ye Li
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Chuanbo Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Hongqi Ai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| |
Collapse
|
33
|
Pérez-Carrión MD, Posadas I, Solera J, Ceña V. LRRK2 and Proteostasis in Parkinson's Disease. Int J Mol Sci 2022; 23:6808. [PMID: 35743250 PMCID: PMC9224256 DOI: 10.3390/ijms23126808] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Parkinson's disease is a neurodegenerative condition initially characterized by the presence of tremor, muscle stiffness and impaired balance, with the deposition of insoluble protein aggregates in Lewy's Bodies the histopathological hallmark of the disease. Although different gene variants are linked to Parkinson disease, mutations in the Leucine-Rich Repeat Kinase 2 (LRRK2) gene are one of the most frequent causes of Parkinson's disease related to genetic mutations. LRRK2 toxicity has been mainly explained by an increase in kinase activity, but alternative mechanisms have emerged as underlying causes for Parkinson's disease, such as the imbalance in LRRK2 homeostasis and the involvement of LRRK2 in aggregation and spreading of α-synuclein toxicity. In this review, we recapitulate the main LRRK2 pathological mutations that contribute to Parkinson's disease and the different cellular and therapeutic strategies devised to correct LRRK2 homeostasis. In this review, we describe the main cellular control mechanisms that regulate LRRK2 folding and aggregation, such as the chaperone network and the protein-clearing pathways such as the ubiquitin-proteasome system and the autophagic-lysosomal pathway. We will also address the more relevant strategies to modulate neurodegeneration in Parkinson's disease through the regulation of LRRK2, using small molecules or LRRK2 silencing.
Collapse
Affiliation(s)
- María Dolores Pérez-Carrión
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; (M.D.P.-C.); (I.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Consorcio CIBER, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Inmaculada Posadas
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; (M.D.P.-C.); (I.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Consorcio CIBER, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Solera
- Servicio de Medicina Interna, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain;
- Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; (M.D.P.-C.); (I.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Consorcio CIBER, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
34
|
PQBP1: The Key to Intellectual Disability, Neurodegenerative Diseases, and Innate Immunity. Int J Mol Sci 2022; 23:ijms23116227. [PMID: 35682906 PMCID: PMC9180999 DOI: 10.3390/ijms23116227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
The idea that a common pathology underlies various neurodegenerative diseases and dementias has attracted considerable attention in the basic and medical sciences. Polyglutamine binding protein-1 (PQBP1) was identified in 1998 after a molecule was predicted to bind to polyglutamine tract amino acid sequences, which are associated with a family of neurodegenerative disorders called polyglutamine diseases. Hereditary gene mutations of PQBP1 cause intellectual disability, whereas acquired loss of function of PQBP1 contributes to dementia pathology. PQBP1 functions in innate immune cells as an intracellular receptor that recognizes pathogens and neurodegenerative proteins. It is an intrinsically disordered protein that generates intracellular foci, similar to other neurodegenerative disease proteins such as TDP43, FUS, and hnRNPs. The knowledge accumulated over more than 20 years has given rise to a new concept that shifts in the equilibrium between physiological and pathological processes have their basis in the dysregulation of common protein structure-linked molecular mechanisms.
Collapse
|
35
|
Affinity of aromatic amino acid side chains in amino acid solvents. Biophys Chem 2022; 287:106831. [DOI: 10.1016/j.bpc.2022.106831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/21/2022] [Accepted: 05/22/2022] [Indexed: 11/17/2022]
|
36
|
Constructing conformational library for amyloid-β42 dimers as the smallest toxic oligomers using two CHARMM force fields. J Mol Graph Model 2022; 115:108207. [DOI: 10.1016/j.jmgm.2022.108207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/19/2022]
|
37
|
Dual-Functional Antioxidant and Antiamyloid Cerium Oxide Nanoparticles Fabricated by Controlled Synthesis in Water-Alcohol Solutions. Biomedicines 2022; 10:biomedicines10050942. [PMID: 35625679 PMCID: PMC9138294 DOI: 10.3390/biomedicines10050942] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/08/2022] [Accepted: 04/17/2022] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress is known to be associated with a number of degenerative diseases. A better knowledge of the interplay between oxidative stress and amyloidogenesis is crucial for the understanding of both, aging and age-related neurodegenerative diseases. Cerium dioxide nanoparticles (CeO2 NPs, nanoceria) due to their remarkable properties are perspective nanomaterials in the study of the processes accompanying oxidative-stress-related diseases, including amyloid-related pathologies. In the present work, we analyze the effects of CeO2 NPs of different sizes and Ce4+/Ce3+ ratios on the fibrillogenesis of insulin, SOD-like enzymatic activity, oxidative stress, biocompatibility, and cell metabolic activity. CeO2 NPs (marked as Ce1–Ce5) with controlled physical–chemical parameters, such as different sizes and various Ce4+/Ce3+ ratios, are synthesized by precipitation in water–alcohol solutions. All synthesized NPs are monodispersed and exhibit good stability in aqueous suspensions. ThT and ANS fluorescence assays and AFM are applied to monitor the insulin amyloid aggregation and antiamyloid aggregation activity of CeO2 NPs. The analyzed Ce1–Ce5 nanoparticles strongly inhibit the formation of insulin amyloid aggregates in vitro. The bioactivity is analyzed using SOD and MTT assays, Western blot, fluorescence microscopy, and flow cytometry. The antioxidative effects and bioactivity of nanoparticles are size- or valence-dependent. CeO2 NPs show great potential benefits for studying the interplay between oxidative stress and amyloid-related diseases, and can be used for verification of the role of oxidative stress in amyloid-related diseases.
Collapse
|
38
|
Alpaugh M, Masnata M, de Rus Jacquet A, Lepinay E, Denis HL, Saint-Pierre M, Davies P, Planel E, Cicchetti F. Passive immunization against phosphorylated tau improves features of Huntington's disease pathology. Mol Ther 2022; 30:1500-1522. [PMID: 35051614 PMCID: PMC9077324 DOI: 10.1016/j.ymthe.2022.01.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 01/07/2023] Open
Abstract
Huntington's disease is classically described as a neurodegenerative disorder of monogenic aetiology. The disease is characterized by an abnormal polyglutamine expansion in the huntingtin gene, which drives the toxicity of the mutated form of the protein. However, accumulation of the microtubule-associated protein tau, which is involved in a number of neurological disorders, has also been observed in patients with Huntington's disease. In order to unravel the contribution of tau hyperphosphorylation to hallmark features of Huntington's disease, we administered weekly intraperitoneal injections of the anti-tau pS202 CP13 monoclonal antibody to zQ175 mice and characterized the resulting behavioral and biochemical changes. After 12 weeks of treatment, motor impairments, cognitive performance and general health were improved in zQ175 mice along with a significant reduction in hippocampal pS202 tau levels. Despite the lack of effect of CP13 on neuronal markers associated with Huntington's disease pathology, tau-targeting enzymes and gliosis, CP13 was shown to directly impact mutant huntingtin aggregation such that brain levels of amyloid fibrils and huntingtin oligomers were decreased, while larger huntingtin protein aggregates were increased. Investigation of CP13 treatment of Huntington's disease patient-derived induced pluripotent stem cells (iPSCs) revealed a reduction in pS202 levels in differentiated cortical neurons and a rescue of neurite length. Collectively, these findings suggest that attenuating tau pathology could mitigate behavioral and molecular hallmarks associated with Huntington's disease.
Collapse
Affiliation(s)
- Melanie Alpaugh
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Maria Masnata
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Aurelie de Rus Jacquet
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Eva Lepinay
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Hélèna L Denis
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Martine Saint-Pierre
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Peter Davies
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Emmanuel Planel
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada.
| |
Collapse
|
39
|
Cujia KS, Herb K, Zopes J, Abendroth JM, Degen CL. Parallel detection and spatial mapping of large nuclear spin clusters. Nat Commun 2022; 13:1260. [PMID: 35273190 PMCID: PMC8913684 DOI: 10.1038/s41467-022-28935-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/08/2021] [Indexed: 11/09/2022] Open
Abstract
Nuclear magnetic resonance imaging (MRI) at the atomic scale offers exciting prospects for determining the structure and function of individual molecules and proteins. Quantum defects in diamond have recently emerged as a promising platform towards reaching this goal, and allowed for the detection and localization of single nuclear spins under ambient conditions. Here, we present an efficient strategy for extending imaging to large nuclear spin clusters, fulfilling an important requirement towards a single-molecule MRI technique. Our method combines the concepts of weak quantum measurements, phase encoding and simulated annealing to detect three-dimensional positions from many nuclei in parallel. Detection is spatially selective, allowing us to probe nuclei at a chosen target radius while avoiding interference from strongly-coupled proximal nuclei. We demonstrate our strategy by imaging clusters containing more than 20 carbon-13 nuclear spins within a radius of 2.4 nm from single, near-surface nitrogen-vacancy centers at room temperature. The radius extrapolates to 5-6 nm for 1H. Beside taking an important step in nanoscale MRI, our experiment also provides an efficient tool for the characterization of large nuclear spin registers in the context of quantum simulators and quantum network nodes.
Collapse
Affiliation(s)
- K S Cujia
- Department of Physics, ETH Zurich, Otto Stern Weg 1, 8093, Zurich, Switzerland. .,IT'IS Foundation, Zeughausstrasse 43, 8004, Zurich, Switzerland.
| | - K Herb
- Department of Physics, ETH Zurich, Otto Stern Weg 1, 8093, Zurich, Switzerland.
| | - J Zopes
- Department of Physics, ETH Zurich, Otto Stern Weg 1, 8093, Zurich, Switzerland. .,Ansys Switzerland GmbH, Technoparkstrasse 1, 8005, Zurich, Switzerland.
| | - J M Abendroth
- Department of Physics, ETH Zurich, Otto Stern Weg 1, 8093, Zurich, Switzerland.
| | - C L Degen
- Department of Physics, ETH Zurich, Otto Stern Weg 1, 8093, Zurich, Switzerland. .,Quantum Center, ETH Zurich, 8093, Zurich, Switzerland.
| |
Collapse
|
40
|
Pavan M, Bassani D, Bolcato G, Bissaro M, Sturles M, Moro S. Computational strategies to identify new drug candidates against neuroinflammation. Curr Med Chem 2022; 29:4756-4775. [PMID: 35135446 DOI: 10.2174/0929867329666220208095122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022]
Abstract
The even more increasing application of computational approaches in these last decades has deeply modified the process of discovery and commercialization of new therapeutic entities. This is especially true in the field of neuroinflammation, in which both the peculiar anatomical localization and the presence of the blood-brain barrier makeit mandatory to finely tune the candidates' physicochemical properties from the early stages of the discovery pipeline. The aim of this review is therefore to provide a general overview to the readers about the topic of neuroinflammation, together with the most common computational strategies that can be exploited to discover and design small molecules controlling neuroinflammation, especially those based on the knowledge of the three-dimensional structure of the biological targets of therapeutic interest. The techniques used to describe the molecular recognition mechanisms, such as molecular docking and molecular dynamics, will therefore be eviscerated, highlighting their advantages and their limitations. Finally, we report several case studies in which computational methods have been applied in drug discovery on neuroinflammation, focusing on the last decade's research.
Collapse
Affiliation(s)
- Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Davide Bassani
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Giovanni Bolcato
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Maicol Bissaro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Mattia Sturles
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences University of Padova, via Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
41
|
Ruifang E, Shi Y, Wang W, Qi M. Callistephin inhibits amyloid-β protein aggregation and determined cytotoxicity against cerebrovascular smooth muscle cells as an in vitro model of cerebral amyloid angiopathy. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
42
|
Tak YJ, Kang S. The E2 ubiquitin-conjugating enzyme HIP2 is a crucial regulator of quality control against mutant SOD1 proteotoxicity. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166316. [PMID: 34856358 DOI: 10.1016/j.bbadis.2021.166316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022]
Abstract
Mutations in superoxide dismutase 1 (SOD1) leading to the formation of intracellular protein aggregates cause amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder characterized by a selective degeneration of motor neurons. The ALS-linked mutant SOD1 emerged as a possible target for ubiquitin-proteasome system (UPS)-mediated degradation. We aimed to elucidate the role of huntingtin interaction protein 2 (HIP2), an E2 ubiquitin-conjugating enzyme, in the proteotoxicity of mutant SOD1 aggregates. We found that HIP2 interacts with mutant SOD1, but not wild-type SOD1, and is upregulated in response to mutant SOD1 expression. Upregulation of HIP2 protein was observed in the spinal cord of 16-week-old SOD1-G93A transgenic mice. HIP2 further modified mutant SOD1 proteins via K48-linked polyubiquitination and degraded mutant SOD1 proteins through the UPS. Upregulation of HIP2 protected cells from mutant SOD1-induced toxicity. Taken together, our findings demonstrate that HIP2 is a crucial regulator of quality control against the proteotoxicity of mutant SOD1. Our results suggest that modulating HIP2 may represent a novel therapeutic strategy for the treatment of ALS.
Collapse
Affiliation(s)
- Yeong Jin Tak
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Seongman Kang
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
43
|
The Use of Bioactive Compounds in Hyperglycemia- and Amyloid Fibrils-Induced Toxicity in Type 2 Diabetes and Alzheimer’s Disease. Pharmaceutics 2022; 14:pharmaceutics14020235. [PMID: 35213966 PMCID: PMC8879577 DOI: 10.3390/pharmaceutics14020235] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/29/2022] Open
Abstract
It has become increasingly apparent that defective insulin signaling may increase the risk for developing Alzheimer’s disease (AD), influence neurodegeneration through promotion of amyloid formation or by increasing inflammatory responses to intraneuronal β-amyloid. Recent work has demonstrated that hyperglycemia is linked to cognitive decline, with elevated levels of glucose causing oxidative stress in vulnerable tissues such as the brain. The ability of β-amyloid peptide to form β-sheet-rich aggregates and induce apoptosis has made amyloid fibrils a leading target for the development of novel pharmacotherapies used in managing and treatment of neuropathological conditions such as AD-related cognitive decline. Additionally, deposits of β-sheets folded amylin, a glucose homeostasis regulator, are also present in diabetic patients. Thus, therapeutic compounds capable of reducing intracellular protein aggregation in models of neurodegenerative disorders may prove useful in ameliorating type 2 diabetes mellitus symptoms. Furthermore, both diabetes and neurodegenerative conditions, such as AD, are characterized by chronic inflammatory responses accompanied by the presence of dysregulated inflammatory biomarkers. This review presents current evidence describing the role of various small bioactive molecules known to ameliorate amyloidosis and subsequent effects in prevention and development of diabetes and AD. It also highlights the potential efficacy of peptide–drug conjugates capable of targeting intracellular targets.
Collapse
|
44
|
Shah S, Dooms MM, Amaral-Garcia S, Igoillo-Esteve M. Current Drug Repurposing Strategies for Rare Neurodegenerative Disorders. Front Pharmacol 2022; 12:768023. [PMID: 34992533 PMCID: PMC8724568 DOI: 10.3389/fphar.2021.768023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Rare diseases are life-threatening or chronically debilitating low-prevalent disorders caused by pathogenic mutations or particular environmental insults. Due to their high complexity and low frequency, important gaps still exist in their prevention, diagnosis, and treatment. Since new drug discovery is a very costly and time-consuming process, leading pharmaceutical companies show relatively low interest in orphan drug research and development due to the high cost of investments compared to the low market return of the product. Drug repurposing–based approaches appear then as cost- and time-saving strategies for the development of therapeutic opportunities for rare diseases. In this article, we discuss the scientific, regulatory, and economic aspects of the development of repurposed drugs for the treatment of rare neurodegenerative disorders with a particular focus on Huntington’s disease, Friedreich’s ataxia, Wolfram syndrome, and amyotrophic lateral sclerosis. The role of academia, pharmaceutical companies, patient associations, and foundations in the identification of candidate compounds and their preclinical and clinical evaluation will also be discussed.
Collapse
Affiliation(s)
- Sweta Shah
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | |
Collapse
|
45
|
TDP-43 pathology: from noxious assembly to therapeutic removal. Prog Neurobiol 2022; 211:102229. [DOI: 10.1016/j.pneurobio.2022.102229] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/08/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
|
46
|
Gadhe L, Sakunthala A, Mukherjee S, Gahlot N, Bera R, Sawner AS, Kadu P, Maji SK. Intermediates of α-synuclein aggregation: Implications in Parkinson's disease pathogenesis. Biophys Chem 2021; 281:106736. [PMID: 34923391 DOI: 10.1016/j.bpc.2021.106736] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/27/2021] [Accepted: 11/27/2021] [Indexed: 12/11/2022]
Abstract
Cytoplasmic deposition of aberrantly misfolded α-synuclein (α-Syn) is a common feature of synucleinopathies, including Parkinson's disease (PD). However, the precise pathogenic mechanism of α-Syn in synucleinopathies remains elusive. Emerging evidence has suggested that α-Syn may contribute to PD pathogenesis in several ways; wherein the contribution of fibrillar species, for exerting toxicity and disease transmission, cannot be neglected. Further, the oligomeric species could be the most plausible neurotoxic species causing neuronal cell death. However, understanding the structural and molecular insights of these oligomers are very challenging due to the heterogeneity and transient nature of the species. In this review, we discuss the recent advancements in understanding the formation and role of α-Syn oligomers in PD pathogenesis. We also summarize the different types of α-Syn oligomeric species and potential mechanisms to exert neurotoxicity. Finally, we address the possible ways to target α-Syn as a promising approach against PD and the possible future directions.
Collapse
Affiliation(s)
- Laxmikant Gadhe
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Riya Bera
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
47
|
Southard T, Kelly K, Armien AG. Myocardial protein aggregates in pet guinea pigs. Vet Pathol 2021; 59:157-163. [PMID: 34530659 DOI: 10.1177/03009858211042586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A retrospective study of guinea pigs submitted for necropsy revealed intracytoplasmic inclusions in the cardiomyocytes of 26 of 30 animals. The inclusions were found with approximately the same frequency in male and female guinea pigs and were slightly more common in older animals. In most cases, the animals did not have clinical signs or necropsy findings suggestive of heart failure, and the cause of death or reason for euthanasia was attributed to concurrent disease processes. However, the 4 guinea pigs with the highest inclusion body burden all had pulmonary edema, sometimes with intra-alveolar hemosiderin-laden macrophages, suggestive of heart failure. The inclusions were found in both the left and right ventricular myocardium, mainly in the papillary muscles, but were most common in the right ventricular free wall. No inclusions were detected in the atrial myocardium or in skeletal muscle. The inclusions did not stain with Congo red or periodic acid-Schiff. Electron microscopy revealed dense aggregates of disorganized myofilaments and microtubules that displaced and compressed the adjacent organelles. By immunohistochemistry, there was some scattered immunoreactivity for desmin and actin at the periphery of the inclusions and punctate actin reactivity within the aggregates. The inclusions did not react with antibodies to ubiquitin or cardiac myosin, but were variably reactive for alpha B crystallin, a small heat shock chaperone protein. The inclusions were interpreted as evidence of impaired proteostasis.
Collapse
|
48
|
Plectin in the Central Nervous System and a Putative Role in Brain Astrocytes. Cells 2021; 10:cells10092353. [PMID: 34572001 PMCID: PMC8464768 DOI: 10.3390/cells10092353] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Plectin, a high-molecular-mass cytolinker, is abundantly expressed in the central nervous system (CNS). Currently, a limited amount of data about plectin in the CNS prevents us from seeing the complete picture of how plectin affects the functioning of the CNS as a whole. Yet, by analogy to its role in other tissues, it is anticipated that, in the CNS, plectin also functions as the key cytoskeleton interlinking molecule. Thus, it is likely involved in signalling processes, thereby affecting numerous fundamental functions in the brain and spinal cord. Versatile direct and indirect interactions of plectin with cytoskeletal filaments and enzymes in the cells of the CNS in normal physiological and in pathologic conditions remain to be fully addressed. Several pathologies of the CNS related to plectin have been discovered in patients with plectinopathies. However, in view of plectin as an integrator of a cohesive mesh of cellular proteins, it is important that the role of plectin is also considered in other CNS pathologies. This review summarizes the current knowledge of plectin in the CNS, focusing on plectin isoforms that have been detected in the CNS, along with its expression profile and distribution alongside diverse cytoskeleton filaments in CNS cell types. Considering that the bidirectional communication between neurons and glial cells, especially astrocytes, is crucial for proper functioning of the CNS, we place particular emphasis on the known roles of plectin in neurons, and we propose possible roles of plectin in astrocytes.
Collapse
|
49
|
Gupta R, Sahu M, Srivastava D, Tiwari S, Ambasta RK, Kumar P. Post-translational modifications: Regulators of neurodegenerative proteinopathies. Ageing Res Rev 2021; 68:101336. [PMID: 33775891 DOI: 10.1016/j.arr.2021.101336] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
One of the hallmark features in the neurodegenerative disorders (NDDs) is the accumulation of aggregated and/or non-functional protein in the cellular milieu. Post-translational modifications (PTMs) are an essential regulator of non-functional protein aggregation in the pathogenesis of NDDs. Any alteration in the post-translational mechanism and the protein quality control system, for instance, molecular chaperone, ubiquitin-proteasome system, autophagy-lysosomal degradation pathway, enhances the accumulation of misfolded protein, which causes neuronal dysfunction. Post-translational modification plays many roles in protein turnover rate, accumulation of aggregate and can also help in the degradation of disease-causing toxic metabolites. PTMs such as acetylation, glycosylation, phosphorylation, ubiquitination, palmitoylation, SUMOylation, nitration, oxidation, and many others regulate protein homeostasis, which includes protein structure, functions and aggregation propensity. Different studies demonstrated the involvement of PTMs in the regulation of signaling cascades such as PI3K/Akt/GSK3β, MAPK cascade, AMPK pathway, and Wnt signaling pathway in the pathogenesis of NDDs. Further, mounting evidence suggests that targeting different PTMs with small chemical molecules, which acts as an inhibitor or activator, reverse misfolded protein accumulation and thus enhances the neuroprotection. Herein, we briefly discuss the protein aggregation and various domain structures of different proteins involved in the NDDs, indicating critical amino acid residues where PTMs occur. We also describe the implementation and involvement of various PTMs on signaling cascade and cellular processes in NDDs. Lastly, we implement our current understanding of the therapeutic importance of PTMs in neurodegeneration, along with emerging techniques targeting various PTMs.
Collapse
|
50
|
Marcelo A, Koppenol R, de Almeida LP, Matos CA, Nóbrega C. Stress granules, RNA-binding proteins and polyglutamine diseases: too much aggregation? Cell Death Dis 2021; 12:592. [PMID: 34103467 PMCID: PMC8187637 DOI: 10.1038/s41419-021-03873-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023]
Abstract
Stress granules (SGs) are membraneless cell compartments formed in response to different stress stimuli, wherein translation factors, mRNAs, RNA-binding proteins (RBPs) and other proteins coalesce together. SGs assembly is crucial for cell survival, since SGs are implicated in the regulation of translation, mRNA storage and stabilization and cell signalling, during stress. One defining feature of SGs is their dynamism, as they are quickly assembled upon stress and then rapidly dispersed after the stress source is no longer present. Recently, SGs dynamics, their components and their functions have begun to be studied in the context of human diseases. Interestingly, the regulated protein self-assembly that mediates SG formation contrasts with the pathological protein aggregation that is a feature of several neurodegenerative diseases. In particular, aberrant protein coalescence is a key feature of polyglutamine (PolyQ) diseases, a group of nine disorders that are caused by an abnormal expansion of PolyQ tract-bearing proteins, which increases the propensity of those proteins to aggregate. Available data concerning the abnormal properties of the mutant PolyQ disease-causing proteins and their involvement in stress response dysregulation strongly suggests an important role for SGs in the pathogenesis of PolyQ disorders. This review aims at discussing the evidence supporting the existence of a link between SGs functionality and PolyQ disorders, by focusing on the biology of SGs and on the way it can be altered in a PolyQ disease context.
Collapse
Affiliation(s)
- Adriana Marcelo
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
- PhD Program in Biomedial Sciences, Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Faro, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Rebekah Koppenol
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
- PhD Program in Biomedial Sciences, Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Faro, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Carlos A Matos
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Clévio Nóbrega
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal.
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
- Champalimaud Research Program, Champalimaud Center for the Unknown, Lisbon, Portugal.
| |
Collapse
|