Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (27)

Search Parameters:
Keywords = polyQ toxicity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 4809 KiB  
Article
In Vitro Efficacy of PEI-Derived Lipopolymers in Silencing of Toxic Proteins in a Neuronal Model of Huntington’s Disease
by Luis C. Morales, Luv Modi, Saba Abbasi Dezfouli, Amarnath Praphakar Rajendran, Remant Kc, Vaibhavi Kadam, Simonetta Sipione and Hasan Uludağ
Pharmaceutics 2025, 17(6), 726; https://doi.org/10.3390/pharmaceutics17060726 - 30 May 2025
Viewed by 726
Abstract
Background: Huntington’s Disease (HD) is a neurodegenerative disorder caused by an abnormal extension of a CAG repeat stretch located in the exon 1 of the HTT (IT15) gene, leading to production of a mutated and misfolded Huntingtin protein (muHTT) with an abnormally elongated [...] Read more.
Background: Huntington’s Disease (HD) is a neurodegenerative disorder caused by an abnormal extension of a CAG repeat stretch located in the exon 1 of the HTT (IT15) gene, leading to production of a mutated and misfolded Huntingtin protein (muHTT) with an abnormally elongated polyglutamine (polyQ) region. This mutation causes muHTT to oligomerize and aggregate in the brain, particularly in the striatum and cortex, causing alterations in intracellular trafficking, caspase activation, and ganglioside metabolism, ultimately leading to neuronal damage and death and causing signs and symptoms such as chorea and cognitive dysfunction. Currently, there is no available cure for HD patients; hence, there is a strong need to look for effective therapies. Methods: This study aims to investigate the efficacy of siRNA-containing nano-engineered lipopolymers in selectively silencing the HTT expression in a neuronal model expressing a chimeric protein formed by the human mutated exon 1 of the HTT gene, tagged with GFP. Toxicity of lipopolymers was assessed using MTT assay, while efficacy of silencing was monitored using qRT-PCR, as well as Western blotting/flow cytometry. Changes in muHTT-GFP aggregation were observed using fluorescence microscopy and image analyses. Results: Here, we show that engineered lipopolymers can be used as delivery vehicles for specific siRNAs, decreasing the transcription of the mutated gene, as well as the muHTT protein production and aggregation, with Leu-Fect C being the most effective candidate amongst the assessed lipopolymers. Conclusions: Our findings have profound implications for genetic disorder therapies, highlighting the potential of nano-engineered materials for silencing mutant genes and facilitating molecular transfection across cellular barriers. This successful in vitro study paves the way for future in vivo investigations with preclinical models, offering hope for previously considered incurable diseases such as HD. Full article
Show Figures

Figure 1

25 pages, 6758 KiB  
Article
Dopamine Receptor D3 Induces Transient, mTORC1-Dependent Autophagy That Becomes Persistent, AMPK-Mediated, and Neuroprotective in Experimental Models of Huntington’s Disease
by Diego Luis-Ravelo, Felipe Fumagallo-Reading, Alejandro Febles-Casquero, Jonathan Lopez-Fernandez, Daniel J. Marcellino and Tomas Gonzalez-Hernandez
Cells 2025, 14(9), 652; https://doi.org/10.3390/cells14090652 - 29 Apr 2025
Viewed by 794
Abstract
Huntington disease’s (HD) is a neurodegenerative disorder caused by the expansion of a polyglutamine region (PolyQ) within the huntingtin protein (HTT). Mutated huntingtin (mHTT) is cytotoxic, particularly for striatal medium spiny neurons (MSNs), whose degeneration is the hallmark of HD. Autophagy inducers currently [...] Read more.
Huntington disease’s (HD) is a neurodegenerative disorder caused by the expansion of a polyglutamine region (PolyQ) within the huntingtin protein (HTT). Mutated huntingtin (mHTT) is cytotoxic, particularly for striatal medium spiny neurons (MSNs), whose degeneration is the hallmark of HD. Autophagy inducers currently available promote the clearance of toxic proteins. However, due to their low selectivity and the possibility that prolonged autophagy hampers essential processes in unaffected cells, researchers have questioned their benefits in neurodegenerative diseases. Since MSNs express dopamine receptors D2 (DRD2) and D3 (DRD3) and DRD2/DRD3 agonists may activate autophagy, here, we explored how healthy and mHTT-challenged cells respond to prolonged DRD2/DRD3 agonist treatment. Autophagy activation and its effects on mHTT/polyQ clearance were studied in R6/1 mice (a genetic model of HD), their wild-type littermates, and DRD2- and DRD3-HEK cells expressing a pathogenic (Q74) and a non-pathogenic (Q23) polyQ fragment of mHTT treated with the DRD2/DRD3 agonist pramipexole. Two forms of DRD3-mediated autophagy were found: a transient mTORC1-dependent in WT mice and Q23-DRD3-HEK cells and a persistent AMPK-ULK1-activated in R6/1 mice and Q74-DRD3-HEK cells. This also promoted a robust clearance of soluble mHTT/polyQ and neuroprotection in striatal neurons and DRD3-HEK cells. The findings indicate that DRD3-induced autophagy may be a safe, disease-modifying intervention in HD patients. Full article
(This article belongs to the Special Issue Molecular Therapeutic Advances for Neurodegenerative Diseases)
Show Figures

Figure 1

14 pages, 2005 KiB  
Article
Quantification of Hydrogen Peroxide in PVP and PVPVA Using 1H qNMR Spectroscopy
by Isha Saraf, Varun Kushwah, Bernd Werner, Klaus Zangger and Amrit Paudel
Polymers 2025, 17(6), 739; https://doi.org/10.3390/polym17060739 - 11 Mar 2025
Cited by 1 | Viewed by 1105
Abstract
Objective: Peroxides in pharmaceutical products and excipients pose risks by oxidizing drug molecules, leading to potential toxicity and reduced efficacy. Accurate peroxide quantification is essential to ensure product safety and potency. This study explores the use of quantitative proton nuclear magnetic resonance ( [...] Read more.
Objective: Peroxides in pharmaceutical products and excipients pose risks by oxidizing drug molecules, leading to potential toxicity and reduced efficacy. Accurate peroxide quantification is essential to ensure product safety and potency. This study explores the use of quantitative proton nuclear magnetic resonance (1H qNMR) spectroscopy as a sensitive and specific method for quantifying peroxide levels in pharmaceutical excipients. Methods: 1H qNMR spectroscopy was employed to measure peroxide levels down to 0.1 ppm in excipients, focusing on poly(vinylpyrrolidone) (PVP) and polyvinylpyrrolidone/vinyl acetate (PVPVA). Different grades and vendors were analyzed, and the impact of various manufacturing processes on hydrogen peroxide content was examined. Results: Peroxide levels varied among different grades of PVP and PVPVA, as well as between vendors. Furthermore, manufacturing processes influenced the hydrogen peroxide content in selected excipients. These variations highlight the importance of controlling peroxide levels in raw materials and during production. Conclusions: 1H qNMR spectroscopy is a valuable tool for accurately quantifying peroxide levels in pharmaceutical excipients. The study emphasizes the need for regular monitoring of peroxide content to ensure the stability, quality, and safety of excipients and drug products. Accurate peroxide measurement can prevent oxidative degradation, preserving both the safety and efficacy of pharmaceutical formulations. Full article
(This article belongs to the Special Issue Polymers for Drug/Gene Delivery and Controlled Release)
Show Figures

Figure 1

13 pages, 1915 KiB  
Article
Erucin, a Natural Isothiocyanate, Prevents Polyglutamine-Induced Toxicity in Caenorhabditis elegans via aak-2/AMPK and daf-16/FOXO Signaling
by Martina Balducci, Julia Tortajada Pérez, Cristina Trujillo del Río, Mar Collado Pérez, Andrea del Valle Carranza, Ana Pilar Gomez Escribano, Rafael P. Vázquez-Manrique and Andrea Tarozzi
Int. J. Mol. Sci. 2024, 25(22), 12220; https://doi.org/10.3390/ijms252212220 - 14 Nov 2024
Cited by 3 | Viewed by 1491
Abstract
Several neurodegenerative diseases (NDDs), such as Huntington’s disease, six of the spinocerebellar ataxias, dentatorubral-pallidoluysian atrophy, and spinobulbar muscular atrophy, are caused by abnormally long polyglutamine (polyQ) tracts. Natural compounds capable of alleviating polyQ-induced toxicity are currently of great interest. In this work, we [...] Read more.
Several neurodegenerative diseases (NDDs), such as Huntington’s disease, six of the spinocerebellar ataxias, dentatorubral-pallidoluysian atrophy, and spinobulbar muscular atrophy, are caused by abnormally long polyglutamine (polyQ) tracts. Natural compounds capable of alleviating polyQ-induced toxicity are currently of great interest. In this work, we investigated the modulatory effect against polyQ neurotoxic aggregates exerted by erucin (ERN), an isothiocyanate naturally present in its precursor glucoerucin in rocket salad leaves and in its oxidized form, sulforaphane (SFN), in broccoli. Using C. elegans models expressing polyQ in different tissues, we demonstrated that ERN protects against polyQ-induced toxicity and that its action depends on the catalytic subunit of AMP-activated protein kinase (aak-2/AMPKα2) and, downstream in this pathway, on the daf-16/FOXO transcription factor, since nematodes deficient in aak-2/AMPKα2 and daf-16 did not respond to the treatment, respectively. Although triggered by a different source of neurotoxicity than polyQ diseases, i.e., by α-synuclein (α-syn) aggregates, Parkinson’s disease (PD) was also considered in our study. Our results showed that ERN reduces α-syn aggregates and slightly improves the motility of worms. Therefore, further preclinical studies in mouse models of protein aggregation are justified and could provide insights into testing whether ERN could be a potential neuroprotective compound in humans. Full article
Show Figures

Graphical abstract

17 pages, 2259 KiB  
Article
Nanoparticle-Mediated Delivery of Deferasirox: A Promising Strategy Against Invasive Aspergillosis
by Sydney Peppe, Moloud Farrokhi, Evan A. Waite, Mustafa Muhi and Efthymia Iliana Matthaiou
Bioengineering 2024, 11(11), 1115; https://doi.org/10.3390/bioengineering11111115 - 5 Nov 2024
Viewed by 1618
Abstract
Background: Invasive aspergillosis (IA) is a deadly fungal lung infection. Antifungal resistance and treatment side effects are major concerns. Iron chelators are vital for IA management, but systemic use can cause side effects. We developed nanoparticles (NPs) to selectively deliver the iron chelator [...] Read more.
Background: Invasive aspergillosis (IA) is a deadly fungal lung infection. Antifungal resistance and treatment side effects are major concerns. Iron chelators are vital for IA management, but systemic use can cause side effects. We developed nanoparticles (NPs) to selectively deliver the iron chelator deferasirox (DFX) for IA treatment. Methods: DFX was encapsulated in poly(lactic-co-glycolic acid) (PLGA) NPs using a single emulsion solvent evaporation method. The NPs were characterized by light scattering and electron microscopy. DFX loading efficiency and release were assessed spectrophotometrically. Toxicity was evaluated using SRB, luciferase, and XTT assays. Therapeutic efficacy was tested in an IA mouse model, assessing fungal burden by qPCR and biodistribution via imaging. Results: DFX-NPs had a size of ~50 nm and a charge of ~−30 mV, with a loading efficiency of ~80%. Release kinetics showed DFX release via diffusion and bioerosion. The EC50 of DFX-NPs was significantly lower (p < 0.001) than the free drug, and they were significantly less toxic (p < 0.0001) in mammalian cell cultures. In vivo, NP treatment significantly reduced Af burden (p < 0.05). Conclusion: The designed DFX-NPs effectively target and kill Af with minimal toxicity to mammalian cells. The significant in vivo therapeutic efficacy suggests these NPs could be a safe and effective treatment for IA. Full article
(This article belongs to the Special Issue Nano–Bio Interface)
Show Figures

Graphical abstract

14 pages, 2588 KiB  
Article
UBL3 Interacts with PolyQ-Expanded Huntingtin Fragments and Modifies Their Intracellular Sorting
by Soho Oyama, Hengsen Zhang, Rafia Ferdous, Yuna Tomochika, Bin Chen, Shuyun Jiang, Md. Shoriful Islam, Md. Mahmudul Hasan, Qing Zhai, A. S. M. Waliullah, Yashuang Ping, Jing Yan, Mst. Afsana Mimi, Chi Zhang, Shuhei Aramaki, Yusuke Takanashi, Tomoaki Kahyo, Yoshio Hashizume, Daita Kaneda and Mitsutoshi Setou
Neurol. Int. 2024, 16(6), 1175-1188; https://doi.org/10.3390/neurolint16060089 - 22 Oct 2024
Viewed by 1917
Abstract
Background/Objectives: UBL3 (Ubiquitin-like 3) is a protein that plays a crucial role in post-translational modifications, particularly in regulating protein transport within small extracellular vesicles. While previous research has predominantly focused on its interactions with α-synuclein, this study investigates UBL3’s role in Huntington’s disease [...] Read more.
Background/Objectives: UBL3 (Ubiquitin-like 3) is a protein that plays a crucial role in post-translational modifications, particularly in regulating protein transport within small extracellular vesicles. While previous research has predominantly focused on its interactions with α-synuclein, this study investigates UBL3’s role in Huntington’s disease (HD). HD is characterized by movement disorders and cognitive impairments, with its pathogenesis linked to toxic, polyglutamine (polyQ)-expanded mutant huntingtin fragments (mHTT). However, the mechanisms underlying the interaction between UBL3 and mHTT remain poorly understood. Methods: To elucidate this relationship, we performed hematoxylin and eosin (HE) staining and immunohistochemistry (IHC) on postmortem brain tissue from HD patients. Gaussia princeps-based split-luciferase complementation assay and co-immunoprecipitation were employed to confirm the interaction between UBL3 and mHTT. Additionally, we conducted a HiBiT lytic detection assay to assess the influence of UBL3 on the intracellular sorting of mHTT. Finally, immunocytochemical staining was utilized to validate the colocalization and distribution of these proteins. Results: Our findings revealed UBL3-positive inclusions in the cytoplasm and nuclei of neurons throughout the striatum of HD patients. We discovered that UBL3 colocalizes and interacts with mHTT and modulates its intracellular sorting. Conclusions: These results suggest that UBL3 may play a significant role in the interaction and sorting of mHTT, contributing to the understanding of its potential implications in the pathophysiology of Huntington’s disease. Full article
(This article belongs to the Special Issue New Insights into Genetic Neurological Diseases)
Show Figures

Figure 1

49 pages, 2110 KiB  
Review
Reviewing the Structure–Function Paradigm in Polyglutamine Disorders: A Synergistic Perspective on Theoretical and Experimental Approaches
by Nastasia Sanda Moldovean-Cioroianu
Int. J. Mol. Sci. 2024, 25(12), 6789; https://doi.org/10.3390/ijms25126789 - 20 Jun 2024
Cited by 3 | Viewed by 2084
Abstract
Polyglutamine (polyQ) disorders are a group of neurodegenerative diseases characterized by the excessive expansion of CAG (cytosine, adenine, guanine) repeats within host proteins. The quest to unravel the complex diseases mechanism has led researchers to adopt both theoretical and experimental methods, each offering [...] Read more.
Polyglutamine (polyQ) disorders are a group of neurodegenerative diseases characterized by the excessive expansion of CAG (cytosine, adenine, guanine) repeats within host proteins. The quest to unravel the complex diseases mechanism has led researchers to adopt both theoretical and experimental methods, each offering unique insights into the underlying pathogenesis. This review emphasizes the significance of combining multiple approaches in the study of polyQ disorders, focusing on the structure–function correlations and the relevance of polyQ-related protein dynamics in neurodegeneration. By integrating computational/theoretical predictions with experimental observations, one can establish robust structure–function correlations, aiding in the identification of key molecular targets for therapeutic interventions. PolyQ proteins’ dynamics, influenced by their length and interactions with other molecular partners, play a pivotal role in the polyQ-related pathogenic cascade. Moreover, conformational dynamics of polyQ proteins can trigger aggregation, leading to toxic assembles that hinder proper cellular homeostasis. Understanding these intricacies offers new avenues for therapeutic strategies by fine-tuning polyQ kinetics, in order to prevent and control disease progression. Last but not least, this review highlights the importance of integrating multidisciplinary efforts to advancing research in this field, bringing us closer to the ultimate goal of finding effective treatments against polyQ disorders. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Neurobiology 2024)
Show Figures

Figure 1

21 pages, 2574 KiB  
Article
AAV-Mediated CAG-Targeting Selectively Reduces Polyglutamine-Expanded Protein and Attenuates Disease Phenotypes in a Spinocerebellar Ataxia Mouse Model
by Anna Niewiadomska-Cimicka, Lorraine Fievet, Magdalena Surdyka, Ewelina Jesion, Céline Keime, Elisabeth Singer, Aurélie Eisenmann, Zaneta Kalinowska-Poska, Hoa Huu Phuc Nguyen, Agnieszka Fiszer, Maciej Figiel and Yvon Trottier
Int. J. Mol. Sci. 2024, 25(8), 4354; https://doi.org/10.3390/ijms25084354 - 15 Apr 2024
Cited by 8 | Viewed by 2598
Abstract
Polyglutamine (polyQ)-encoding CAG repeat expansions represent a common disease-causing mutation responsible for several dominant spinocerebellar ataxias (SCAs). PolyQ-expanded SCA proteins are toxic for cerebellar neurons, with Purkinje cells (PCs) being the most vulnerable. RNA interference (RNAi) reagents targeting transcripts with expanded CAG reduce [...] Read more.
Polyglutamine (polyQ)-encoding CAG repeat expansions represent a common disease-causing mutation responsible for several dominant spinocerebellar ataxias (SCAs). PolyQ-expanded SCA proteins are toxic for cerebellar neurons, with Purkinje cells (PCs) being the most vulnerable. RNA interference (RNAi) reagents targeting transcripts with expanded CAG reduce the level of various mutant SCA proteins in an allele-selective manner in vitro and represent promising universal tools for treating multiple CAG/polyQ SCAs. However, it remains unclear whether the therapeutic targeting of CAG expansion can be achieved in vivo and if it can ameliorate cerebellar functions. Here, using a mouse model of SCA7 expressing a mutant Atxn7 allele with 140 CAGs, we examined the efficacy of short hairpin RNAs (shRNAs) targeting CAG repeats expressed from PHP.eB adeno-associated virus vectors (AAVs), which were introduced into the brain via intravascular injection. We demonstrated that shRNAs carrying various mismatches with the CAG target sequence reduced the level of polyQ-expanded ATXN7 in the cerebellum, albeit with varying degrees of allele selectivity and safety profile. An shRNA named A4 potently reduced the level of polyQ-expanded ATXN7, with no effect on normal ATXN7 levels and no adverse side effects. Furthermore, A4 shRNA treatment improved a range of motor and behavioral parameters 23 weeks after AAV injection and attenuated the disease burden of PCs by preventing the downregulation of several PC-type-specific genes. Our results show the feasibility of the selective targeting of CAG expansion in the cerebellum using a blood–brain barrier-permeable vector to attenuate the disease phenotype in an SCA mouse model. Our study represents a significant advancement in developing CAG-targeting strategies as a potential therapy for SCA7 and possibly other CAG/polyQ SCAs. Full article
Show Figures

Figure 1

12 pages, 1893 KiB  
Review
Role of TFEB in Huntington’s Disease
by Javier Ojalvo-Pacheco, Sokhna M. S. Yakhine-Diop, José M. Fuentes, Marta Paredes-Barquero and Mireia Niso-Santano
Biology 2024, 13(4), 238; https://doi.org/10.3390/biology13040238 - 4 Apr 2024
Viewed by 3308
Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease caused by an expansion of the CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. This expansion leads to a polyglutamine (polyQ) tract at the N-terminal end of HTT, which reduces the [...] Read more.
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease caused by an expansion of the CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. This expansion leads to a polyglutamine (polyQ) tract at the N-terminal end of HTT, which reduces the solubility of the protein and promotes its accumulation. Inefficient clearance of mutant HTT (mHTT) by the proteasome or autophagy–lysosomal system leads to accumulation of oligomers and toxic protein aggregates in neurons, resulting in impaired proteolytic systems, transcriptional dysregulation, impaired axonal transport, mitochondrial dysfunction and cellular energy imbalance. Growing evidence suggests that the accumulation of mHTT aggregates and autophagic and/or lysosomal dysfunction are the major pathogenic mechanisms underlying HD. In this context, enhancing autophagy may be an effective therapeutic strategy to remove protein aggregates and improve cell function. Transcription factor EB (TFEB), a master transcriptional regulator of autophagy, controls the expression of genes critical for autophagosome formation, lysosomal biogenesis, lysosomal function and autophagic flux. Consequently, the induction of TFEB activity to promote intracellular clearance may be a therapeutic strategy for HD. However, while some studies have shown that overexpression of TFEB facilitates the clearance of mHTT aggregates and ameliorates the disease phenotype, others indicate such overexpression may lead to mHTT co-aggregation and worsen disease progression. Further studies are necessary to confirm whether TFEB modulation could be an effective therapeutic strategy against mHTT-mediated toxicity in different disease models. Full article
(This article belongs to the Special Issue Lysosomes and Diseases Associated with Its Dysfunction)
Show Figures

Figure 1

14 pages, 1977 KiB  
Review
Spinocerebellar Ataxia Type 3 Pathophysiology—Implications for Translational Research and Clinical Studies
by Fabian Stahl, Bernd O. Evert, Xinyu Han, Peter Breuer and Ullrich Wüllner
Int. J. Mol. Sci. 2024, 25(7), 3984; https://doi.org/10.3390/ijms25073984 - 3 Apr 2024
Cited by 2 | Viewed by 3901
Abstract
The spinocerebellar ataxias (SCA) comprise a group of inherited neurodegenerative diseases. Machado–Joseph Disease (MJD) or spinocerebellar ataxia 3 (SCA3) is the most common autosomal dominant form, caused by the expansion of CAG repeats within the ataxin-3 (ATXN3) gene. This mutation results in the [...] Read more.
The spinocerebellar ataxias (SCA) comprise a group of inherited neurodegenerative diseases. Machado–Joseph Disease (MJD) or spinocerebellar ataxia 3 (SCA3) is the most common autosomal dominant form, caused by the expansion of CAG repeats within the ataxin-3 (ATXN3) gene. This mutation results in the expression of an abnormal protein containing long polyglutamine (polyQ) stretches that confers a toxic gain of function and leads to misfolding and aggregation of ATXN3 in neurons. As a result of the neurodegenerative process, SCA3 patients are severely disabled and die prematurely. Several screening approaches, e.g., druggable genome-wide and drug library screenings have been performed, focussing on the reduction in stably overexpressed ATXN3(polyQ) protein and improvement in the resultant toxicity. Transgenic overexpression models of toxic ATXN3, however, missed potential modulators of endogenous ATXN3 regulation. In another approach to identify modifiers of endogenous ATXN3 expression using a CRISPR/Cas9-modified SK-N-SH wild-type cell line with a GFP-T2A-luciferase (LUC) cassette under the control of the endogenous ATXN3 promotor, four statins were identified as potential activators of expression. We here provide an overview of the high throughput screening approaches yet performed to find compounds or genomic modifiers of ATXN3(polyQ) toxicity in different SCA3 model organisms and cell lines to ameliorate and halt SCA3 progression in patients. Furthermore, the putative role of cholesterol in neurodegenerative diseases (NDDs) in general and SCA3 in particular is discussed. Full article
(This article belongs to the Special Issue CNS Drug Action in Neurodegenerative Diseases 3.0)
Show Figures

Figure 1

16 pages, 856 KiB  
Review
Towards Understanding Neurodegenerative Diseases: Insights from Caenorhabditis elegans
by Yingjie Wu, Yining Chen, Xiaochun Yu, Minxing Zhang and Zhaoyu Li
Int. J. Mol. Sci. 2024, 25(1), 443; https://doi.org/10.3390/ijms25010443 - 28 Dec 2023
Cited by 8 | Viewed by 3988
Abstract
The elevated occurrence of debilitating neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), Alzheimer’s disease (AD), Parkinson’s disease (PD) and Machado–Joseph disease (MJD), demands urgent disease-modifying therapeutics. Owing to the evolutionarily conserved molecular signalling pathways with mammalian species and facile [...] Read more.
The elevated occurrence of debilitating neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), Alzheimer’s disease (AD), Parkinson’s disease (PD) and Machado–Joseph disease (MJD), demands urgent disease-modifying therapeutics. Owing to the evolutionarily conserved molecular signalling pathways with mammalian species and facile genetic manipulation, the nematode Caenorhabditis elegans (C. elegans) emerges as a powerful and manipulative model system for mechanistic insights into neurodegenerative diseases. Herein, we review several representative C. elegans models established for five common neurodegenerative diseases, which closely simulate disease phenotypes specifically in the gain-of-function aspect. We exemplify applications of high-throughput genetic and drug screenings to illustrate the potential of C. elegans to probe novel therapeutic targets. This review highlights the utility of C. elegans as a comprehensive and versatile platform for the dissection of neurodegenerative diseases at the molecular level. Full article
(This article belongs to the Special Issue C. elegans as a Disease Model: Molecular Perspectives)
Show Figures

Figure 1

9 pages, 3433 KiB  
Communication
Treatment with the Glycosphingolipid Modulator THI Rescues Myelin Integrity in the Striatum of R6/2 HD Mice
by Giuseppe Pepe, Paola Lenzi, Luca Capocci, Federico Marracino, Ludovica Pizzati, Pamela Scarselli, Alba Di Pardo, Francesco Fornai and Vittorio Maglione
Int. J. Mol. Sci. 2023, 24(6), 5956; https://doi.org/10.3390/ijms24065956 - 22 Mar 2023
Cited by 1 | Viewed by 2065
Abstract
Huntington’s disease is one of the most common dominantly inherited neurodegenerative disorders caused by an expansion of a polyglutamine (polyQ) stretch in the N-terminal region of huntingtin (Htt). Among all the molecular mechanisms, affected by the mutation, emerging evidence proposes glycosphingolipid dysfunction as [...] Read more.
Huntington’s disease is one of the most common dominantly inherited neurodegenerative disorders caused by an expansion of a polyglutamine (polyQ) stretch in the N-terminal region of huntingtin (Htt). Among all the molecular mechanisms, affected by the mutation, emerging evidence proposes glycosphingolipid dysfunction as one of the major determinants. High levels of sphingolipids have been found to localize in the myelin sheaths of oligodendrocytes, where they play an important role in myelination stability and functions. In this study, we investigated any potential existing link between sphingolipid modulation and myelin structure by performing both ultrastructural and biochemical analyses. Our findings demonstrated that the treatment with the glycosphingolipid modulator THI preserved myelin thickness and the overall structure and reduced both area and diameter of pathologically giant axons in the striatum of HD mice. These ultrastructural findings were associated with restoration of different myelin marker protein, such as myelin-associated glycoprotein (MAG), myelin basic protein (MBP) and 2′, 3′ Cyclic Nucleotide 3′-Phosphodiesterase (CNP). Interestingly, the compound modulated the expression of glycosphingolipid biosynthetic enzymes and increased levels of GM1, whose elevation has been extensively reported to be associated with reduced toxicity of mutant Htt in different HD pre-clinical models. Our study further supports the evidence that the metabolism of glycosphingolipids may represent an effective therapeutic target for the disease. Full article
(This article belongs to the Special Issue Huntington’s Disease: Molecular Advances in Therapeutics)
Show Figures

Figure 1

20 pages, 363 KiB  
Review
What the Gut Tells the Brain—Is There a Link between Microbiota and Huntington’s Disease?
by Dorota Wronka, Anna Karlik, Julia O. Misiorek and Lukasz Przybyl
Int. J. Mol. Sci. 2023, 24(5), 4477; https://doi.org/10.3390/ijms24054477 - 24 Feb 2023
Cited by 20 | Viewed by 5013
Abstract
The human intestinal microbiota is a diverse and dynamic microenvironment that forms a complex, bi-directional relationship with the host. The microbiome takes part in the digestion of food and the generation of crucial nutrients such as short chain fatty acids (SCFA), but is [...] Read more.
The human intestinal microbiota is a diverse and dynamic microenvironment that forms a complex, bi-directional relationship with the host. The microbiome takes part in the digestion of food and the generation of crucial nutrients such as short chain fatty acids (SCFA), but is also impacts the host’s metabolism, immune system, and even brain functions. Due to its indispensable role, microbiota has been implicated in both the maintenance of health and the pathogenesis of many diseases. Dysbiosis in the gut microbiota has already been implicated in many neurodegenerative diseases such as Parkinson’s disease (PD) and Alzheimer’s disease (AD). However, not much is known about the microbiome composition and its interactions in Huntington’s disease (HD). This dominantly heritable, incurable neurodegenerative disease is caused by the expansion of CAG trinucleotide repeats in the huntingtin gene (HTT). As a result, toxic RNA and mutant protein (mHTT), rich in polyglutamine (polyQ), accumulate particularly in the brain, leading to its impaired functions. Interestingly, recent studies indicated that mHTT is also widely expressed in the intestines and could possibly interact with the microbiota, affecting the progression of HD. Several studies have aimed so far to screen the microbiota composition in mouse models of HD and find out whether observed microbiome dysbiosis could affect the functions of the HD brain. This review summarizes ongoing research in the HD field and highlights the essential role of the intestine-brain axis in HD pathogenesis and progression. The review also puts a strong emphasis on indicating microbiome composition as a future target in the urgently needed therapy for this still incurable disease. Full article
(This article belongs to the Special Issue Molecular Research in Human Microbiome)
Show Figures

Graphical abstract

16 pages, 1226 KiB  
Review
Mechanistic Insights and Potential Therapeutic Approaches in PolyQ Diseases via Autophagy
by Mukul Jain, Nil Patil, Gholamreza Abdi, Maryam Abbasi Tarighat, Arifullah Mohammed, Muhammad Rajaei Ahmad Mohd Zain and Khang Wen Goh
Biomedicines 2023, 11(1), 162; https://doi.org/10.3390/biomedicines11010162 - 9 Jan 2023
Cited by 5 | Viewed by 3550
Abstract
Polyglutamine diseases are a group of congenital neurodegenerative diseases categorized with genomic abnormalities in the expansion of CAG triplet repeats in coding regions of specific disease-related genes. Protein aggregates are the toxic hallmark for polyQ diseases and initiate neuronal death. Autophagy is a [...] Read more.
Polyglutamine diseases are a group of congenital neurodegenerative diseases categorized with genomic abnormalities in the expansion of CAG triplet repeats in coding regions of specific disease-related genes. Protein aggregates are the toxic hallmark for polyQ diseases and initiate neuronal death. Autophagy is a catabolic process that aids in the removal of damaged organelles or toxic protein aggregates, a process required to maintain cellular homeostasis that has the potential to fight against neurodegenerative diseases, but this pathway gets affected under diseased conditions, as there is a direct impact on autophagy-related gene expression. The increase in the accumulation of autophagy vesicles reported in neurodegenerative diseases was due to an increase in autophagy or may have been due to a decrease in autophagy flux. These reports suggested that there is a contribution of autophagy in the pathology of diseases and regulation in the process of autophagy. It was demonstrated in various disease models of polyQ diseases that autophagy upregulation by using modulators can enhance the dissolution of toxic aggregates and delay disease progression. In this review, interaction of the autophagy pathway with polyQ diseases was analyzed, and a therapeutic approach with autophagy inducing drugs was established for disease pathogenesis. Full article
Show Figures

Figure 1

13 pages, 5891 KiB  
Article
Adsorption of Diclofenac Sodium by Aged Degradable and Non-Degradable Microplastics: Environmental Effects, Adsorption Mechanisms
by Siqi Liang, Kangkang Wang, Kefu Wang, Yuli Kou, Tao Wang, Changyan Guo, Wei Wang and Jide Wang
Toxics 2023, 11(1), 24; https://doi.org/10.3390/toxics11010024 - 27 Dec 2022
Cited by 18 | Viewed by 4084
Abstract
Microplastics (MPs) are novel pollutants, which can carry toxic contaminants and are released in biota and accumulate. The adsorption behavior of MPs and aged MPs has attracted extensive attention. In this paper, the aging process of polystyrene (PS) and poly (butyleneadipate-co-terephthalate) (PBAT) plastics [...] Read more.
Microplastics (MPs) are novel pollutants, which can carry toxic contaminants and are released in biota and accumulate. The adsorption behavior of MPs and aged MPs has attracted extensive attention. In this paper, the aging process of polystyrene (PS) and poly (butyleneadipate-co-terephthalate) (PBAT) plastics under ultraviolet (UV) irradiation at a high temperature and their adsorption properties for the contaminant diclofenac sodium (DCF) before and after aging was investigated. There are many factors affecting the adsorption capacity of MPs. In this experiment, three aspects of MPs, organic pollutants, and environmental factors are explored. The Freundlich model as well as the pseudosecondary kinetic model is more applicable to the process of DCF adsorption by MPs. The main effects of adsorption of organic pollutants by MPs are electrostatic interactions, hydrogen-halogen bonds, and hydrophobic interactions. The adsorption capacity of the UV-aged MPs on DCF is significantly enhanced, and the order of adsorption capacity is Q(A-PBAT) (27.65 mg/g) > Q (A-PS) (23.91 mg/g) > Q (PBAT) (9.30 mg/g) > Q (PS) (9.21 mg/g). The results show that more active sites are generated on the surface of MPs after aging, which can enhance their adsorption capacity for organic pollutants. This adsorption mechanism will increase their role as contaminant carriers in the aquatic food chain. Full article
(This article belongs to the Section Toxicity Reduction and Environmental Remediation)
Show Figures

Figure 1

Back to TopTop