1
|
Cheng J, Koch ET, Ramandi D, Mackay JP, O'Leary TP, Rees-Jones W, Raymond LA. Synaptic modulation of glutamate in striatum of the YAC128 mouse model of Huntington disease. Neurobiol Dis 2025; 205:106774. [PMID: 39716682 DOI: 10.1016/j.nbd.2024.106774] [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: 10/02/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Altered balance between striatal direct and indirect pathways contributes to early motor, cognitive and psychiatric symptoms in Huntington disease (HD). While degeneration of striatal D2-type dopamine receptor (D2)-expressing indirect pathway medium spiny neurons (iMSNs) occurs prior to that of D1-type dopamine receptor (D1)-expressing direct pathway neurons, altered corticostriatal synaptic function precedes degeneration. D2-mediated signaling on iMSNs reduces their excitability and promotes endocannabinoid (eCB) synthesis, suppressing glutamate release from cortical afferents. D2 receptors are also expressed on glutamatergic cortical terminals, cholinergic interneurons, and dopaminergic terminals from substantia nigra where they suppress release of glutamate, acetylcholine and dopamine, respectively, and these cell types may contribute to early striatal dysfunction in HD. Thus, we used corticostriatal brain slices and optogenetic probes to directly investigate neuromodulatory signaling in the transgenic YAC128 HD mouse model. RESULTS Low-dose D2 agonist quinpirole reduced cortically-evoked glutamate release in dorsal striatum of premanifest YAC128 slices but not WT, and blocking type 1 cannabinoid receptors mitigated this effect. YAC128 corticostriatal brain slices also showed increased evoked dopamine and reduced evoked eCB release compared to WT, while acetylcholine signaling patterns remained relatively intact. CONCLUSIONS These findings suggest that YAC128 corticostriatal slices show increased D2 sensitivity that is eCB-dependent, and that dopamine and eCB release are altered at an early disease stage. We provide evidence for impaired neuromodulatory signaling in early HD, guiding therapeutic efforts prior to the onset of overt motor symptoms later on.
Collapse
Affiliation(s)
- Judy Cheng
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Ellen T Koch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Daniel Ramandi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Cell and Developmental Biology, University of British Columbia, Vancouver, BC, Canada
| | - James P Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Timothy P O'Leary
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - William Rees-Jones
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
2
|
Wu F, Luo H, Wang X, Yang Q, Zhuang Y, Lin L, Dong Y, Tulupov A, Zhang Y, Cai S, Chen Z, Cai C, Bao J, Cheng J. Application of Anti-Motion Ultra-Fast Quantitative MRI in Neurological Disorder Imaging: Insights From Huntington's Disease. J Magn Reson Imaging 2025. [PMID: 39887812 DOI: 10.1002/jmri.29682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Conventional quantitative MRI (qMRI) scan is time-consuming and highly sensitive to movements, posing great challenges for quantitative images of individuals with involuntary movements, such as Huntington's disease (HD). PURPOSE To evaluate the potential of our developed ultra-fast qMRI technique, multiple overlapping-echo detachment (MOLED), in overcoming involuntary head motion and its capacity to quantitatively assess tissue changes in HD. STUDY TYPE Prospective. PHANTOM/SUBJECTS A phantom comprising 13 tubes of MnCl2 at varying concentrations, 5 healthy volunteers (male/female: 1/4), 22 HD patients (male/female: 14/8) and 27 healthy controls (male/female: 15/12). FIELD STRENGTH/SEQUENCE 3.0 T. MOLED-T2 sequence, MOLED-T2* sequence, T2-weighted spin-echo sequence, T1-weighted gradient echo sequence, and T2-dark-fluid sequence. ASSESSMENT T1-weighted images were reconstructed into high-resolution images, followed by segmentation to delineate regions of interest (ROIs). Subsequently, the MOLED T2 and T2* maps were aligned with the high-resolution images, and the ROIs were transformed into the MOLED image space using the transformation matrix and warp field. Finally, T2 and T2* values were extracted from the MOLED relaxation maps. STATISTICAL TESTS Bland-Altman analysis, independent t test, Mann-Whitney U test, Pearson correlation analysis, and Spearman correlation analysis, P < 0.05 was considered statistically significant. RESULTS MOLED-T2 and MOLED-T2* sequences demonstrated good accuracy (Meandiff = - 0.20%, SDdiff = 1.05%, and Meandiff = -1.73%, SDdiff = 10.98%, respectively), and good repeatability (average intraclass correlation coefficient: 0.856 and 0.853, respectively). More important, MOLED T2 and T2* maps remained artifact-free across all HD patients, even in the presence of apparent head motions. Moreover, there were significant differences in T2 and T2* values across multiple ROIs between HD and controls. DATA CONCLUSION The ultra-fast scanning capabilities of MOLED effectively mitigate the impact of head movements, offering a robust solution for quantitative imaging in HD. Moreover, T2 and T2* values derived from MOLED provide powerful capabilities for quantifying tissue changes. PLAIN LANGUAGE SUMMARY Quantitative MRI scan is time-consuming and sensitive to movements. Consequently, obtaining quantitative images is challenging for patients with involuntary movements, such as those with Huntington's Disease (HD). In response, a newly developed MOLED technique has been introduced, promising to resist motion through ultra-fast scan. This technique has demonstrated excellent accuracy and reproducibility and importantly all HD patient's MOLED maps remained artifacts-free. Additionally, there were significant differences in T2 and T2∗ values across ROIs between HD and controls. The robust resistance of MOLED to motion makes it particularly suitable for quantitative assessments in patients prone to involuntary movements. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Fei Wu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Haiyang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiao Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Qinqin Yang
- Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, Xiamen University, Xiamen, China
| | - Yuchuan Zhuang
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Liangjie Lin
- Clinical and technical support, Philips Healthcare, Beijing, China
| | - Yanbo Dong
- Institute of Psychology, The Herzen State Pedagogical University of Russia, Saint Petersburg, Russia
| | - Andrey Tulupov
- Laboratory of MRT technologies, The Institute International Tomography Center of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuhui Cai
- Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, Xiamen University, Xiamen, China
| | - Zhong Chen
- Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, Xiamen University, Xiamen, China
| | - Congbo Cai
- Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, Xiamen University, Xiamen, China
| | - Jianfeng Bao
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Giannoula A, De Paepe AE, Sanz F, Furlong LI, Camara E. Identifying time patterns in Huntington's disease trajectories using dynamic time warping-based clustering on multi-modal data. Sci Rep 2025; 15:3081. [PMID: 39856140 PMCID: PMC11759715 DOI: 10.1038/s41598-025-86686-5] [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: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
One of the principal goals of Precision Medicine is to stratify patients by accounting for individual variability. However, extracting meaningful information from Real-World Data, such as Electronic Health Records, still remains challenging due to methodological and computational issues. A Dynamic Time Warping-based unsupervised-clustering methodology is presented in this paper for the clustering of patient trajectories of multi-modal health data on the basis of shared temporal characteristics. Building on an earlier methodology, a new dimension of time-varying clinical and imaging features is incorporated, through an adapted cost-minimization algorithm for clustering on different, possibly overlapping, feature subsets. The model disease chosen is Huntington's disease (HD), characterized by progressive neurodegeneration. From a wide range of examined user-defined parameters, four case examples are highlighted to demonstrate the identified temporal patterns in multi-modal HD trajectories and to study how these differ due to the combined effects of feature weights and granularity threshold. For each identified cluster, polynomial fits that describe the time behavior of the assessed features are provided for an informative comparison, together with their averaged values. The proposed data-mining methodology permits the stratification of distinct time patterns of multi-modal health data in individuals that share a diagnosis, by employing user-customized criteria beyond the current clinical practice. Overall, this work bears implications for better analysis of individual variability in disease progression, opening doors to personalized preventative, diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Alexia Giannoula
- Research Group on Integrative Biomedical Informatics (GRIB), Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Hospital del Mar Research Institute, Barcelona, Spain.
| | - Audrey E De Paepe
- Research Group on Integrative Biomedical Informatics (GRIB), Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Hospital del Mar Research Institute, Barcelona, Spain
- Cognition and Brain Plasticity Unit, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Ferran Sanz
- Research Group on Integrative Biomedical Informatics (GRIB), Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Estela Camara
- Cognition and Brain Plasticity Unit, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| |
Collapse
|
4
|
Joshi DC, Chavan MB, Gurow K, Gupta M, Dhaliwal JS, Ming LC. The role of mitochondrial dysfunction in Huntington's disease: Implications for therapeutic targeting. Biomed Pharmacother 2025; 183:117827. [PMID: 39854819 DOI: 10.1016/j.biopha.2025.117827] [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: 10/11/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Huntington's disease (HD) is a progressive, autosomal dominant neurodegenerative disorder characterized by cognitive decline, motor dysfunction, and psychiatric disturbances. A common feature of neurodegenerative disorders is mitochondrial dysfunction, which affects the brain's sensitivity to oxidative damage and its high oxygen demand. This dysfunction may plays a significant role in the pathogenesis of Huntington's disease. HD is caused by a CAG repeat expansion in the huntingtin gene, which leads to the production of a toxic mutant huntingtin (mHTT) protein. This disruption in mitochondrial function compromises energy metabolism and increases oxidative stress, resulting in mitochondrial DNA abnormalities, impaired calcium homeostasis, and altered mitochondrial dynamics. These effects ultimately may contribute to neuronal dysfunction and cell death, underscoring the importance of targeting mitochondrial function in developing therapeutic strategies for HD. This review discusses the mechanistic role of mitochondrial dysfunction in Huntington's disease. Mitochondrial dysfunction is a crucial factor in HD, making mitochondrial-targeted therapies a promising approach for treatment. We explore therapies that address bioenergy deficits, antioxidants that reduce reactive oxygen species, calcium modulators that restore calcium homeostasis, and treatments that enhance mitochondrial dynamics to rejuvenate mitochondrial function. We also highlight innovative treatment approaches such as gene editing and stem cell therapy, which offer hope for more personalized strategies. In conclusion, understanding mitochondrial dysfunction in Huntington's disease may guide potential treatment strategies. Targeting this dysfunction may help to slow disease progression and enhance the quality of life for individuals affected by Huntington's disease.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Mayuri Bapu Chavan
- TMV's Lokmanya Tilak Institute of Pharmaceutical Sciences, Pune, Maharashtra, India.
| | - Kajal Gurow
- Department of Pharmacology, Gurukul Pharmacy college, Ranpur, Kota, Rajasthan, India
| | - Madhu Gupta
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India.
| | | | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (deemed to be University), Sawangi (M), Wardha, India.
| |
Collapse
|
5
|
Tuan J. Diagnosing Huntington's disease on the medical ward. BMJ Case Rep 2025; 18:e264004. [PMID: 39828326 DOI: 10.1136/bcr-2024-264004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
An African American man in his early 40s with progressive gait impairment and chronic cognitive impairment initially presented to the emergency department after statements of self-harm and was hospitalised. Examination revealed notable neurological abnormalities including impaired memory recall, oral dyskinesia/choreiform movements, dystonia of the right upper extremity with drift, hyper-reflexia and spastic gait. On further evaluation, including neurology and genetics consultation and workup, a clinical diagnosis of the neurodegenerative disorder Huntington's disease (HD) was made. Further history revealed a family history of cognitive issues and dystonia in his uncle at the age of mid-40s. The diagnosis of HD was confirmed via genetic testing of the blood, specifically looking for trinucleotide repeats. HD allele 1 had full penetrance with 44 cytosine-adenine-guanine (CAG) repeats and HD allele 2 had partial penetrance with 15 CAG repeats. A multidisciplinary team was critical in diagnosing and managing this patient's underlying HD.
Collapse
Affiliation(s)
- Jessica Tuan
- Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
6
|
Tian L, Qiang T, Liu S, Zhang B, Zhang Y, Zhang B, Hu J, Zhang J, Lu Q, Ke C, Xia J, Liang C. Cannabinoid receptor 1 ligands: Biased signaling mechanisms driving functionally selective drug discovery. Pharmacol Ther 2025; 267:108795. [PMID: 39828030 DOI: 10.1016/j.pharmthera.2025.108795] [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: 09/05/2024] [Revised: 12/20/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
G protein-coupled receptors (GPCRs) adopt conformational states that activate or inhibit distinct signaling pathways, including those mediated by G proteins or β-arrestins. Biased signaling through GPCRs may offer a promising strategy to enhance therapeutic efficacy while reducing adverse effects. Cannabinoid receptor 1 (CB1), a key GPCR in the endocannabinoid system, presents therapeutic potential for conditions such as pain, anxiety, cognitive impairment, psychiatric disorders, and metabolic diseases. This review examines the structural conformations of CB1 coupling to different signaling pathways and explores the mechanisms underlying biased signaling, which are critical for the design of functionally selective ligands. We discuss the structure-function relationships of endogenous cannabinoids (eCBs), phytocannabinoids, and synthetic cannabinoid ligands with biased properties. Challenges such as the complexity of ligand bias screening, the limited availability of distinctly biased ligands, and the variability in receptor signaling profiles in vivo have hindered clinical progress. Although the therapeutic potential of biased ligands in various clinical conditions remains in its infancy, retrospective identification of such molecules provides a strong foundation for further development. Recent advances in CB1 crystallography, particularly insights into its conformations with G proteins and β-arrestins, now offer a framework for structure-based drug design. While there is still a long way to go before biased CB1 ligands can be widely used in clinical practice, ongoing multidisciplinary research shows promise for achieving functional selectivity in targeting specific pathways. These progress could lead to the development of safer and more effective cannabinoid-based therapies in the future.
Collapse
Affiliation(s)
- Lei Tian
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China; Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Taotao Qiang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Sundian Liu
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Boxin Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Yunfei Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Bingxing Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jinrong Hu
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jiayun Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Qi Lu
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Changhua Ke
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Juan Xia
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524045, China
| | - Chengyuan Liang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Shaanxi University of Science & Technology, Xi'an 710021, China; School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China.
| |
Collapse
|
7
|
Kapplingattu SV, Bhattacharya S, Adlakha YK. MiRNAs as major players in brain health and disease: current knowledge and future perspectives. Cell Death Discov 2025; 11:7. [PMID: 39805813 PMCID: PMC11729916 DOI: 10.1038/s41420-024-02283-x] [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: 10/09/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
MicroRNAs are regulators of gene expression and their dysregulation can lead to various diseases. MicroRNA-135 (MiR-135) exhibits brain-specific expression, and performs various functions such as neuronal morphology, neural induction, and synaptic function in the human brain. Dysfunction of miR-135 has been reported in brain tumors, and neurodegenerative and neurodevelopmental disorders. Several reports show downregulation of miR-135 in glioblastoma, indicating its tumor suppressor role in the pathogenesis of brain tumors. In this review, by performing in silico analysis of molecular targets of miR-135, we reveal the significant pathways and processes modulated by miR-135. We summarize the biological significance, roles, and signaling pathways of miRNAs in general, with a focus on miR-135 in different neurological diseases including brain tumors, and neurodegenerative and neurodevelopmental disorders. We also discuss methods, limitations, and potential of glioblastoma organoids in recapitulating disease initiation and progression. We highlight the promising therapeutic potential of miRNAs as antitumor agents for aggressive human brain tumors including glioblastoma.
Collapse
Affiliation(s)
- Sarika V Kapplingattu
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Yogita K Adlakha
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India.
| |
Collapse
|
8
|
Delint-Ramirez I, Madabhushi R. DNA damage and its links to neuronal aging and degeneration. Neuron 2025; 113:7-28. [PMID: 39788088 DOI: 10.1016/j.neuron.2024.12.001] [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: 09/12/2024] [Revised: 11/07/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
DNA damage is a major risk factor for the decline of neuronal functions with age and in neurodegenerative diseases. While how DNA damage causes neurodegeneration is still being investigated, innovations over the past decade have provided significant insights into this issue. Breakthroughs in next-generation sequencing methods have begun to reveal the characteristics of neuronal DNA damage hotspots and the causes of DNA damage. Chromosome conformation capture-based approaches have shown that, while DNA damage and the ensuing cellular response alter chromatin topology, chromatin organization at damage sites also affects DNA repair outcomes in neurons. Additionally, neuronal activity results in the formation of programmed DNA breaks, which could burden DNA repair mechanisms and promote neuronal dysfunction. Finally, emerging evidence implicates DNA damage-induced inflammation as an important contributor to the age-related decline in neuronal functions. Together, these discoveries have ushered in a new understanding of the significance of genome maintenance for neuronal function.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- Departments of Psychiatry, Neuroscience, and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O' Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ram Madabhushi
- Departments of Psychiatry, Neuroscience, and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O' Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
9
|
Alves F, Lane D, Nguyen TPM, Bush AI, Ayton S. In defence of ferroptosis. Signal Transduct Target Ther 2025; 10:2. [PMID: 39746918 PMCID: PMC11696223 DOI: 10.1038/s41392-024-02088-5] [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: 06/24/2024] [Revised: 10/10/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Rampant phospholipid peroxidation initiated by iron causes ferroptosis unless this is restrained by cellular defences. Ferroptosis is increasingly implicated in a host of diseases, and unlike other cell death programs the physiological initiation of ferroptosis is conceived to occur not by an endogenous executioner, but by the withdrawal of cellular guardians that otherwise constantly oppose ferroptosis induction. Here, we profile key ferroptotic defence strategies including iron regulation, phospholipid modulation and enzymes and metabolite systems: glutathione reductase (GR), Ferroptosis suppressor protein 1 (FSP1), NAD(P)H Quinone Dehydrogenase 1 (NQO1), Dihydrofolate reductase (DHFR), retinal reductases and retinal dehydrogenases (RDH) and thioredoxin reductases (TR). A common thread uniting all key enzymes and metabolites that combat lipid peroxidation during ferroptosis is a dependence on a key cellular reductant, nicotinamide adenine dinucleotide phosphate (NADPH). We will outline how cells control central carbon metabolism to produce NADPH and necessary precursors to defend against ferroptosis. Subsequently we will discuss evidence for ferroptosis and NADPH dysregulation in different disease contexts including glucose-6-phosphate dehydrogenase deficiency, cancer and neurodegeneration. Finally, we discuss several anti-ferroptosis therapeutic strategies spanning the use of radical trapping agents, iron modulation and glutathione dependent redox support and highlight the current landscape of clinical trials focusing on ferroptosis.
Collapse
Affiliation(s)
- Francesca Alves
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Darius Lane
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
10
|
Wu J, Santos-Garcia I, Eiriz I, Brüning T, Kvasnička A, Friedecký D, Nyman TA, Pahnke J. Sex-dependent efficacy of sphingosine-1-phosphate receptor agonist FTY720 in mitigating Huntington's disease. Pharmacol Res 2025; 211:107557. [PMID: 39725338 DOI: 10.1016/j.phrs.2024.107557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative disorder characterized by severe motor deficits, cognitive decline and psychiatric disturbances. An early and significant morphological hallmark of HD is the activation of astrocytes triggered by mutant huntingtin, leading to the release of inflammatory mediators. Fingolimod (FTY), an FDA-approved sphingosine-1-phosphate (S1P) receptor agonist is used to treat multiple sclerosis (MS), a neuroinflammatory disease, and has shown therapeutic promise in other neurological conditions. Our study aimed to investigate the therapeutic potential of FTY for treating HD by utilizing a well-characterized mouse model of HD (zQ175dn) and wild-type littermates. The study design included a crossover, long-term oral treatment with 1 mg/kg to 2 mg/kg FTY from the age of 15-46 weeks (n = 128). Different motor behavior and physiological parameters were assessed throughout the study. The findings revealed that FTY rescued disease-related body weight loss in a sex-dependent manner, indicating its potential to regulate metabolic disturbances and to counteract neurodegenerative processes in HD. FTY intervention also rescued testicular atrophy, restored testis tissue structure in male mice suggesting a broader impact on peripheral tissues affected by huntingtin pathology. Histological analyses of the brain revealed delayed accumulation of activated astrocytes contributing to the preservation of the neural microenvironment by reducing neuroinflammation. The extent of FTY-related disease improvement was sex-dependent. Motor functions and body weight improved mostly in female mice with sustained estrogen levels, whereas males had to compensate for the ongoing, disease-related testis atrophy and the loss of androgen production. Our study underscores the beneficial therapeutic effects of FTY on HD involving endogenous steroid hormones and their important anabolic effects. It positions FTY as a promising candidate for therapeutic interventions targeting various aspects of HD pathology. Further studies are needed to fully evaluate its therapeutic potential in patients.
Collapse
Affiliation(s)
- Jingyun Wu
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo N-0372, Norway
| | - Irene Santos-Garcia
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo N-0372, Norway
| | - Ivan Eiriz
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo N-0372, Norway
| | - Thomas Brüning
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo N-0372, Norway
| | - Aleš Kvasnička
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, Zdravotníků 248/7, Olomouc CZ-77900, Czech Republic
| | - David Friedecký
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, Zdravotníků 248/7, Olomouc CZ-77900, Czech Republic
| | - Tuula A Nyman
- Proteomics Core Facility (PCF), Department of Immunology, Oslo University Hospital (OUS) and University of Oslo (UiO), Faculty of Medicine, Sognsvannsveien 20, Oslo NO-0372, Norway
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo N-0372, Norway; Institute of Nutritional Medicine (INUM) and Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, Lübeck D-23538, Germany; Department of Neuromedicine and Neuroscience, Faculty of Medicine and Life Sciences, University of Latvia (LU), Jelgavas iela 3, Rīga LV-1004, Latvia; Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Ramat Aviv IL-6997801, Israel.
| |
Collapse
|
11
|
Kumar U. Cannabinoids: Role in Neurological Diseases and Psychiatric Disorders. Int J Mol Sci 2024; 26:152. [PMID: 39796008 PMCID: PMC11720483 DOI: 10.3390/ijms26010152] [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: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
An impact of legalization and decriminalization of marijuana is the gradual increase in the use of cannabis for recreational purposes, which poses a potential threat to society and healthcare systems worldwide. However, the discovery of receptor subtypes, endogenous endocannabinoids, and enzymes involved in synthesis and degradation, as well as pharmacological characterization of receptors, has led to exploration of the use of cannabis in multiple peripheral and central pathological conditions. The role of cannabis in the modulation of crucial events involving perturbed physiological functions and disease progression, including apoptosis, inflammation, oxidative stress, perturbed mitochondrial function, and the impaired immune system, indicates medicinal values. These events are involved in most neurological diseases and prompt the gradual progression of the disease. At present, several synthetic agonists and antagonists, in addition to more than 70 phytocannabinoids, are available with distinct efficacy as a therapeutic alternative in different pathological conditions. The present review aims to describe the use of cannabis in neurological diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
12
|
Ratovitski T, Holland CD, O’Meally RN, Shevelkin AV, Shi T, Cole RN, Jiang M, Ross CA. Huntingtin interactome reveals huntingtin role in regulation of double strand break DNA damage response (DSB/DDR), chromatin remodeling and RNA processing pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630542. [PMID: 39763784 PMCID: PMC11703178 DOI: 10.1101/2024.12.27.630542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Huntington's Disease (HD), a progressive neurodegenerative disorder with no disease-modifying therapies, is caused by a CAG repeat expansion in the HD gene encoding polyglutamine-expanded huntingtin (HTT) protein. Mechanisms of HD cellular pathogenesis and cellular functions of the normal and mutant HTT proteins are still not completely understood. HTT protein has numerous interaction partners, and it likely provides a scaffold for assembly of multiprotein complexes many of which may be altered in HD. Previous studies have implicated DNA damage response in HD pathogenesis. Gene transcription and RNA processing has also emerged as molecular mechanisms associated with HD. Here we used multiple approaches to identify HTT interactors in the context of DNA damage stress. Our results indicate that HTT interacts with many proteins involved in the regulation of interconnected DNA repair/remodeling and RNA processing pathways. We present evidence for a role for HTT in double strand break repair mechanism. We demonstrate HTT functional interaction with a major DNA damage response kinase DNA-PKcs and association of both proteins with nuclear speckles. We show that S1181 phosphorylation of HTT is regulated by DSB, and can be carried out (at least in vitro) by DNA-PK. Furthermore, we show HTT interactions with RNA binding proteins associated with nuclear speckles, including two proteins encoded by genes at HD modifier loci, TCERG1 and MED15, and with chromatin remodeling complex BAF. These interactions of HTT may position it as an important scaffolding intermediary providing integrated regulation of gene expression and RNA processing in the context of DNA repair mechanisms.
Collapse
Affiliation(s)
- Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 5 South, Baltimore, MD21287
| | - Chloe D. Holland
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 5 South, Baltimore, MD21287
| | - Robert N. O’Meally
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 N. Broadway Street, Suite 371 BRB Baltimore, MD21287
| | - Alexey V. Shevelkin
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 5 South, Baltimore, MD21287
| | - Tianze Shi
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 5 South, Baltimore, MD21287
| | - Robert N. Cole
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 N. Broadway Street, Suite 371 BRB Baltimore, MD21287
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 5 South, Baltimore, MD21287
| | - Christopher A. Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 5 South, Baltimore, MD21287
- Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| |
Collapse
|
13
|
Ahmad SR, Zeyaullah M, Khan MS, AlShahrani AM, Altijani AAG, Ali H, Dawria A, Mohieldin A, Alam MS, Mohamed AOA. Pharmacogenomics for neurodegenerative disorders - a focused review. Front Pharmacol 2024; 15:1478964. [PMID: 39759457 PMCID: PMC11695131 DOI: 10.3389/fphar.2024.1478964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 01/07/2025] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are characterized by the progressive degeneration of neuronal structure and function, leading to severe cognitive and motor impairments. These conditions present significant challenges to healthcare systems, and traditional treatments often fail to account for genetic variability among patients, resulting in inconsistent therapeutic outcomes. Pharmacogenomics aims to tailor medical treatments based on an individual's genetic profile, thereby improving therapeutic efficacy and reducing adverse effects. This focused review explores the genetic factors influencing drug responses in neurodegenerative diseases and the potential of pharmacogenomics to revolutionize their treatment. Key genetic markers, such as the APOE ε4 allele in AD and the CYP2D6 polymorphisms in PD, are highlighted for their roles in modulating drug efficacy. Additionally, advancements in pharmacogenomic tools, including genome-wide association studies (GWAS), next-generation sequencing (NGS), and CRISPR-Cas9, are discussed for their contributions to personalized medicine. The application of pharmacogenomics in clinical practice and its prospects, including ethical and data integration challenges, are also examined.
Collapse
Affiliation(s)
- S. Rehan Ahmad
- Hiralal Mazumdar Memorial College for Women, West Bengal State University, Kolkata, India
| | - Md. Zeyaullah
- Department of Basic Medical Science, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Mohammad Suhail Khan
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Abdullah M. AlShahrani
- Department of Basic Medical Science, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Abdelrhman A. Galaleldin Altijani
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Haroon Ali
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Adam Dawria
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Ali Mohieldin
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| | - Mohammad Shane Alam
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Awad Osman Abdalla Mohamed
- Department of Anaesthesia Technology, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University (KKU), Abha, Saudi Arabia
| |
Collapse
|
14
|
Brzezinski M, Argudo PG, Scheidt T, Yu M, Hosseini E, Kaltbeitzel A, Lemke EA, Michels JJ, Parekh SH. Protein-Specific Crowding Accelerates Aging in Protein Condensates. Biomacromolecules 2024. [PMID: 39648588 DOI: 10.1021/acs.biomac.4c00609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Macromolecular crowding agents, such as poly(ethylene glycol) (PEG), are often used to mimic cellular cytoplasm in protein assembly studies. Despite the perception that crowding agents have an inert nature, we demonstrate and quantitatively explore the diverse effects of PEG on the phase separation and maturation of protein condensates. We use two model proteins, the FG domain of Nup98 and bovine serum albumin (BSA), which represent an intrinsically disordered protein and a protein with a well-established secondary structure, respectively. PEG expedites the maturation of Nup98, enhancing denser protein packing and fortifying interactions, which hasten beta-sheet formation and subsequent droplet gelation. In contrast to BSA, PEG enhances droplet stability and limits the available solvent for protein solubilization, inducing only minimal changes in the secondary structure, pointing toward a significantly different role of the crowding agent. Strikingly, we detect almost no presence of PEG in Nup droplets, whereas PEG is moderately detectable within BSA droplets. Our findings demonstrate a nuanced interplay between crowding agents and proteins; PEG can accelerate protein maturation in liquid-liquid phase separation systems, but its partitioning and effect on protein structure in droplets is protein specific. This suggests that crowding phenomena are specific to each protein-crowding agent pair.
Collapse
Affiliation(s)
- Mateusz Brzezinski
- Department of Biomedical Engineering University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, Texas 78712, United States
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Pablo G Argudo
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Tom Scheidt
- Biocenter, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 17, Mainz 55128, Germany
- Institute of Molecular Biology GmbH, Ackermannweg 4, Mainz 55128, Germany
| | - Miao Yu
- Biocenter, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 17, Mainz 55128, Germany
- Institute of Molecular Biology GmbH, Ackermannweg 4, Mainz 55128, Germany
| | - Elnaz Hosseini
- Department of Biomedical Engineering University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, Texas 78712, United States
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Anke Kaltbeitzel
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Edward A Lemke
- Biocenter, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 17, Mainz 55128, Germany
- Institute of Molecular Biology GmbH, Ackermannweg 4, Mainz 55128, Germany
| | - Jasper J Michels
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Sapun H Parekh
- Department of Biomedical Engineering University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, Texas 78712, United States
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| |
Collapse
|
15
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
16
|
De Paepe AE, Plana-Alcaide Y, Garcia-Gorro C, Rodriguez-Dechicha N, Vaquer I, Calopa M, de Diego-Balaguer R, Camara E. Cognitive engagement may slow clinical progression and brain atrophy in Huntington's disease. Sci Rep 2024; 14:30156. [PMID: 39627260 PMCID: PMC11614872 DOI: 10.1038/s41598-024-76680-8] [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: 06/13/2024] [Accepted: 10/16/2024] [Indexed: 12/06/2024] Open
Abstract
Lifelong cognitive engagement conveys benefits in Huntington's disease (HD) and may positively affect non-cognitive domains in other populations. However, the effect of lifelong cognitive engagement on the progression of motor and psychiatric domains in HD remains unknown, as is its neurobiological basis. Forty-five HD individuals completed the Cognitive Reserve Questionnaire (CRQ) and longitudinal clinical evaluation (maximum total of six visits, mean inter-assessment duration of 13.53 ± 4.1 months). Of these, thirty-three underwent longitudinal neuroimaging (18 ± 6 months follow-up). Generalized linear mixed-effects models were executed to predict the effect of individual differences in lifelong cognitive engagement on HD clinical progression and voxel-based morphometry to explore the impact of lifelong cognitive engagement on whole-brain gray matter volume atrophy. Controlling for age, disease stage, and sex, higher CRQ scores were associated with reduced overall severity and longitudinal progression across cognitive, motor, and psychiatric domains. Those with higher CRQ scores demonstrated reduced gray matter volume loss in the middle frontal gyrus, supplementary motor area, and middle cingulate. This putative impact on HD clinical progression may be conferred by preservation of brain volume in neural hubs that integrate executive function with action initiation and behavioral regulation, providing support for early cognitive engagement, even prior to diagnosis.
Collapse
Grants
- PID2020-114518RB-I00 to EC, BFU2017-87109-P to RdD Agencia Estatal de Investigación (AEI), an agency of MINECO, and co-funded by FEDER funds/European Regional Development Fund (ERDF) - a Way to Build Europe
- PID2020-114518RB-I00 to EC, BFU2017-87109-P to RdD Agencia Estatal de Investigación (AEI), an agency of MINECO, and co-funded by FEDER funds/European Regional Development Fund (ERDF) - a Way to Build Europe
- CP13/00225, PI14/00834 Instituto de Salud Carlos III, which is an agency of the MINECO, co-funded by FEDER funds/European Regional Development Fund (ERDF) - a way to Build Europe
- Agencia Estatal de Investigación (AEI), an agency of MINECO, and co-funded by FEDER funds/European Regional Development Fund (ERDF) – a Way to Build Europe
- Instituto de Salud Carlos III, which is an agency of the MINECO, co-funded by FEDER funds/European Regional Development Fund (ERDF) – a way to Build Europe
Collapse
Affiliation(s)
- Audrey E De Paepe
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute - IDIBELL], L'Hospitalet de Llobregat, Barcelona, 08097, Spain
- Department of Cognition, Development and Education Psychology, Universitat de Barcelona, Barcelona, Spain
| | - Yemila Plana-Alcaide
- Clinical Research of Brain, Cognition and Behavior, Terrassa Health Consortium, Terrassa, Spain
| | - Clara Garcia-Gorro
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute - IDIBELL], L'Hospitalet de Llobregat, Barcelona, 08097, Spain
- Department of Cognition, Development and Education Psychology, Universitat de Barcelona, Barcelona, Spain
| | | | - Irene Vaquer
- Hestia Duran i Reynals. Hospital Duran i Reynals, Hospitalet de Llobregat, Barcelona, Spain
| | - Matilde Calopa
- Movement Disorders Unit, Neurology Service, Bellvitge University Hospital, Barcelona, Spain
| | - Ruth de Diego-Balaguer
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute - IDIBELL], L'Hospitalet de Llobregat, Barcelona, 08097, Spain
- Department of Cognition, Development and Education Psychology, Universitat de Barcelona, Barcelona, Spain
- ICREA (Catalan Institution for Research and Advanced Studies), Barcelona, Spain
| | - Estela Camara
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute - IDIBELL], L'Hospitalet de Llobregat, Barcelona, 08097, Spain.
| |
Collapse
|
17
|
Feleus S, Skotnicki LE, Roos RA, de Bot ST. Medication Use and Treatment Indications in Huntington's Disease; Analyses from a Large Cohort. Mov Disord Clin Pract 2024; 11:1530-1541. [PMID: 39431460 PMCID: PMC11647978 DOI: 10.1002/mdc3.14230] [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: 02/16/2024] [Revised: 07/19/2024] [Accepted: 09/14/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Huntington's Disease is a rare neurodegenerative disorder in which appropriate medication management is essential. While many medications are prescribed based on expert knowledge, overviews of actual medication use in HD are sparse. OBJECTIVES We provide a detailed overview of medication use and associated indications across HD disease stages, considering sex and regional differences. METHODS Data from the largest observational HD study, ENROLL-HD, were used. We created HD-related medication and indication classes to identify medication trends in manifest, premanifest and control subjects. We studied medication use in adult, childhood- and adolescent-onset HD, incorporating disease stage (including phenoconverters), sex and regional differences. RESULTS In 8546 manifest HD patients, 84.6% used medication (any type), with the average number of medications per user rising from 2.5 in premanifest HD to 5.2 in end stage disease. Antipsychotics (29.2%), SSRIs (27.5%) and painkillers (21.8%) were most often used. Medication use varied with disease progression. Several differences were observed between the sexes, and notably between Europe and Northern America as well. Medication use increased after phenoconversion (from 64.8% to 70.6%, P < 0.05), with the largest difference in antipsychotic use (4.4%-7.8%, P < 0.05). Medication patterns were different in childhood-onset HD, with no use of painkillers, less use of anti-chorea and antidepressant drugs, and more for aggression and irritability. CONCLUSIONS Medication use in HD increases with disease progression, with varying types of medications prescribed based on disease stage, sex, and region of living. Recognizing these medication trends is vital for further personalized HD management.
Collapse
Affiliation(s)
- Stephanie Feleus
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Lara E.M. Skotnicki
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | - Raymund A.C. Roos
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | - Susanne T. de Bot
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
18
|
Liu H, Bai Q, Wang X, Jin Y, Ju X, Lu C. Immune signature of gene expression pattern shared by autism spectrum disorder and Huntington's disease. IBRO Neurosci Rep 2024; 17:311-319. [PMID: 39398347 PMCID: PMC11471255 DOI: 10.1016/j.ibneur.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024] Open
Abstract
Autism spectrum disorder (ASD) and Huntington's disease (HD) are complex neurological conditions with unclear causes and limited treatments, affecting individuals, families, and society. Despite ASD and HD representing two opposing stages of neuronal development and degeneration, they share similar clinical-pathological features in motor function. In this study, we leveraged transcriptomic data from the prefrontal cortex available in public databases to identify shared transcriptional characteristics of ASD and HD. Differential expression analysis revealed that the majority of differentially expressed genes (DEGs) were up-regulated in ASD carriers, whereas most DEGs were down-regulated in HD carriers. Among the DEGs shared between both diseases, three out of seven protein-coding genes were related to the immune system. Furthermore, we identified two enriched pathways shared between ASD and HD DEGs. The gene interaction network analysis unveiled four hub genes shared by both diseases, all of which are associated with immune functions. The findings suggest a shared gene expression pattern in the prefrontal cortex of people with ASD and HD, closely linked to the immune system. These findings will contribute to exploring the biological mechanisms underlying the shared phenotypes of these two diseases from an immunological perspective.
Collapse
Affiliation(s)
- Huanhuan Liu
- School of Psychology, Northeast Normal University, Changchun, China
| | - Qiuyu Bai
- Yancheng College of Mechatronic Technology, Yancheng, China
| | | | - Yunlei Jin
- Children’ s Hospital of Changchun, Changchun, China
| | - Xingda Ju
- School of Psychology, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Cognitive Neuroscience and Brain Development, Changchun, China
| | - Chang Lu
- School of Psychology, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Cognitive Neuroscience and Brain Development, Changchun, China
| |
Collapse
|
19
|
Abdian S, Fakhri S, Moradi SZ, Khirehgesh MR, Echeverría J. Saffron and its major constituents against neurodegenerative diseases: A mechanistic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156097. [PMID: 39577115 DOI: 10.1016/j.phymed.2024.156097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Neurodegeneration has been recognized as the main pathophysiological alteration in the majority of brain-related diseases. Despite contemporary attempts to provide acceptable medicinal therapies, the conclusion has not been much beneficial. Besides, the complex pathophysiological mechanisms behind neurodegenerative diseases (NDDs) urge the needs for finding novel multi-target agents. Accordingly, saffron with major active constituents and as multi-targeting agents have shown beneficial effects in modulating NDDs with higher efficacy and lower side effects. PURPOSE The present study provides a systematic and comprehensive review of the existing in vitro, in vivo, and clinical data on the effectiveness, and signaling pathways of saffron and its key phytochemical components in the management of NDDs. The need to develop novel saffron delivery systems is also considered. METHODS Studies were identified through a systematic and comprehensive search in Science Direct, PubMed, and Scopus databases through April 30, 2024. The whole saffron major constituents (e.g., saffron, crocin, crocetin, picrocrocin, and safranal) and NDDs (e.g., neuro*, spinal cord injury, multiple sclerosis, amyotrophic lateral sclerosis, Huntington*, Parkinson*, Alzheimer*, and brain) were selected as keywords to find related studies. In the systematic analysis, 64 articles were directly included in the current study. Additional reports were added within the comprehensive studies in the review. RESULTS Saffron and its active metabolites crocin, crocetin, safranal, and picrocrocin have shown acceptable efficacy in managing NDDs like Alzheimer's disease, Parkinson's disease, Attention deficit hyperactivity disorder, depression, and other NDDs via modulating apoptotic (e.g., caspases, Bax/Bcl-2, cytochrome c, and death receptors), inflammatory (e.g., NF-κB, IL-1β, IL-6, TNF-α, and COX-2), and oxidative strass (e.g., Nrf2, GSH, GPx, CAT, SOD, MDA, ROS, and nitrite) signaling pathways. The presented in vitro, in vivo, and clinical evidences showed us a better future of controlling NDDs with higher efficacy, while decreasing associated side effects with no significant toxicity. Additionally, employing novel delivery systems could increase the efficacy of saffron phytoconstituents to resolve the issues pharmacokinetic limitations. CONCLUSION Saffron and its major constituents employ anti-inflammatory, anti-apoptotic and antioxidant mechanisms in modulating several dysregulated-signaling pathways in NDDs. However, further research is necessary to elucidate the precise underlying mechanisms in exploring the feasibility of using saffron active compounds against NDDs. More studies should focus on dose-response relationships, long-term effects, highlighting key mechanisms, and designing more well-controlled clinical trials. Additionally, developing stable and cost-benefit novel delivery systems in future works helps to remove the pharmacokinetic limitations of saffron major constituents.
Collapse
Affiliation(s)
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Khirehgesh
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
20
|
Gu YY, Zhao XR, Zhang N, Yang Y, Yi Y, Shao QH, Liu MX, Zhang XL. Mitochondrial dysfunction as a therapeutic strategy for neurodegenerative diseases: Current insights and future directions. Ageing Res Rev 2024; 102:102577. [PMID: 39528070 DOI: 10.1016/j.arr.2024.102577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases, as common diseases in the elderly, tend to become younger due to environmental changes, social development and other factors. They are mainly characterized by progressive loss or dysfunction of neurons in the central or peripheral nervous system, and common diseases include Parkinson's disease, Alzheimer's disease, Huntington's disease and so on. Mitochondria are important organelles for adenosine triphosphate (ATP) production in the brain. In recent years, a large amount of evidence has shown that mitochondrial dysfunction plays a direct role in neurodegenerative diseases, which is expected to provide new ideas for the treatment of related diseases. This review will summarize the main mechanisms of mitochondrial dysfunction in neurodegenerative diseases, as well as collating recent advances in the study of mitochondrial disorders and new therapies.
Collapse
Affiliation(s)
- Ying-Ying Gu
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Xin-Ru Zhao
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Nan Zhang
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Yuan Yang
- Department of Gastroenterology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Ying Yi
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Qian-Hang Shao
- Department of Pharmacy, Peking University People's Hospital, Beijing 100871, P R China
| | - Ming-Xuan Liu
- College of Pharmacy, Nantong University, Nantong 226001, PR China.
| | - Xiao-Ling Zhang
- College of Pharmacy, Nantong University, Nantong 226001, PR China.
| |
Collapse
|
21
|
Liu H, McCollum A, Krishnaprakash A, Ouyang Y, Shi T, Ratovitski T, Jiang M, Duan W, Ross CA, Jin J. Roscovitine, a CDK Inhibitor, Reduced Neuronal Toxicity of mHTT by Targeting HTT Phosphorylation at S1181 and S1201 In Vitro. Int J Mol Sci 2024; 25:12315. [PMID: 39596381 PMCID: PMC11594617 DOI: 10.3390/ijms252212315] [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: 10/20/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a single mutation in the huntingtin gene (HTT). Normal HTT has a CAG trinucleotide repeat at its N-terminal within the range of 36. However, once the CAG repeats exceed 37, the mutant gene (mHTT) will encode mutant HTT protein (mHTT), which results in neurodegeneration in the brain, specifically in the striatum and other brain regions. Since the mutation was discovered, there have been many research efforts to understand the mechanism and develop therapeutic strategies to treat HD. HTT is a large protein with many post-translational modification sites (PTMs) and can be modified by phosphorylation, acetylation, methylation, sumoylation, etc. Some modifications reduced mHTT toxicity both in cell and animal models of HD. We aimed to find the known kinase inhibitors that can modulate the toxicity of mHTT. We performed an in vitro kinase assay using HTT peptides, which bear different PTM sites identified by us previously. A total of 368 kinases were screened. Among those kinases, cyclin-dependent kinases (CDKs) affected the serine phosphorylation on the peptides that contain S1181 and S1201 of HTT. We explored the effect of CDK1 and CDK5 on the phosphorylation of these PTMs of HTT and found that CDK5 modified these two serine sites, while CDK5 knockdown reduced the phosphorylation of S1181 and S1201. Modifying these two serine sites altered the neuronal toxicity induced by mHTT. Roscovitine, a CDK inhibitor, reduced the p-S1181 and p-S1201 and had a protective effect against mHTT toxicity. We further investigated the feasibility of the use of roscovitine in HD mice. We confirmed that roscovitine penetrated the mouse brain by IP injection and inhibited CDK5 activity in the brains of HD mice. It is promising to move this study to in vivo for pre-clinical HD treatment.
Collapse
Affiliation(s)
- Hongshuai Liu
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Ainsley McCollum
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Asvini Krishnaprakash
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Yuxiao Ouyang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Tianze Shi
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| | - Christopher A. Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
- Department of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jing Jin
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA (Y.O.); (T.R.); (W.D.); (C.A.R.)
| |
Collapse
|
22
|
King AC, Payne E, Stephens E, Fowler JA, Wood TE, Rodriguez E, Gray M. Modulation of SNARE-dependent exocytosis in astrocytes improves neuropathology in Huntington's disease. Dis Model Mech 2024; 17:dmm052002. [PMID: 39526491 PMCID: PMC11583919 DOI: 10.1242/dmm.052002] [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: 06/20/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Huntington's disease (HD) is a fatal, progressive neurodegenerative disorder. Prior studies revealed an increase in extracellular glutamate levels after evoking astrocytic SNARE-dependent exocytosis from cultured primary astrocytes from mutant huntingtin (mHTT)-expressing BACHD mice compared to control astrocytes, suggesting alterations in astrocytic SNARE-dependent exocytosis in HD. We used BACHD and dominant-negative (dn)SNARE mice to decrease SNARE-dependent exocytosis from astrocytes to determine whether reducing SNARE-dependent exocytosis from astrocytes could rescue neuropathological changes in vivo. We observed significant protection against striatal atrophy and no significant rescue of cortical atrophy in BACHD/dnSNARE mice compared to BACHD mice. Amino acid transporters are important for modulating the levels of extracellular neurotransmitters. BACHD mice had no change in GLT1 expression, decreased striatal GAT1 expression and increased levels of GAT3. There was no change in GAT1 after reducing astrocytic SNARE-dependent exocytosis, and increased GAT3 expression in BACHD mice was normalized in BACHD/dnSNARE mice. Thus, modulation of astrocytic SNARE-dependent exocytosis in BACHD mice is protective against striatal atrophy and modulates GABA transporter expression.
Collapse
Affiliation(s)
- Annesha C. King
- Graduate Biomedical Sciences Neuroscience Theme, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily Payne
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily Stephens
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jahmel A. Fowler
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Graduate Biomedical Sciences Biochemistry and Structural Biology Theme, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tara E. Wood
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Efrain Rodriguez
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michelle Gray
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
23
|
Beura SK, Panigrahi AR, Yadav P, Kulkarni PP, Lakhanpal V, Singh B, Singh SK. Role of Thrombosis in Neurodegenerative Diseases: An Intricate Mechanism of Neurovascular Complications. Mol Neurobiol 2024:10.1007/s12035-024-04589-4. [PMID: 39482419 DOI: 10.1007/s12035-024-04589-4] [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: 04/11/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | | | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Bhupinder Singh
- Department of Cardiology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
24
|
Pupak A, Rodríguez-Navarro I, Sathasivam K, Singh A, Essmann A, Del Toro D, Ginés S, Mouro Pinto R, Bates GP, Vang Ørom UA, Martí E, Brito V. m 6A modification of mutant huntingtin RNA promotes the biogenesis of pathogenic huntingtin transcripts. EMBO Rep 2024; 25:5026-5052. [PMID: 39394467 PMCID: PMC11549361 DOI: 10.1038/s44319-024-00283-7] [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: 12/11/2023] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 10/13/2024] Open
Abstract
In Huntington's disease (HD), aberrant processing of huntingtin (HTT) mRNA produces HTT1a transcripts that encode the pathogenic HTT exon 1 protein. The mechanisms behind HTT1a production are not fully understood. Considering the role of m6A in RNA processing and splicing, we investigated its involvement in HTT1a generation. Here, we show that m6A methylation is increased before the cryptic poly(A) sites (IpA1 and IpA2) within the huntingtin RNA in the striatum of Hdh+/Q111 mice and human HD samples. We further assessed m6A's role in mutant Htt mRNA processing by pharmacological inhibition and knockdown of METTL3, as well as targeted demethylation of Htt intron 1 using a dCas13-ALKBH5 system in HD mouse cells. Our data reveal that Htt1a transcript levels are regulated by both METTL3 and the methylation status of Htt intron 1. They also show that m6A methylation in intron 1 depends on expanded CAG repeats. Our findings highlight a potential role for m6A in aberrant splicing of Htt mRNA.
Collapse
Affiliation(s)
- Anika Pupak
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Irene Rodríguez-Navarro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Kirupa Sathasivam
- Department of Neurodegenerative Disease, Huntington's Disease Centre and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, UCL, London, WC1N 3BG, UK
| | - Ankita Singh
- Department for Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Amelie Essmann
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Daniel Del Toro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Silvia Ginés
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ricardo Mouro Pinto
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gillian P Bates
- Department of Neurodegenerative Disease, Huntington's Disease Centre and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, UCL, London, WC1N 3BG, UK
| | | | - Eulàlia Martí
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Verónica Brito
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
25
|
Li Y, Fu J, Wang H. Advancements in Targeting Ion Channels for the Treatment of Neurodegenerative Diseases. Pharmaceuticals (Basel) 2024; 17:1462. [PMID: 39598374 PMCID: PMC11597607 DOI: 10.3390/ph17111462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/20/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Ion channels are integral membrane proteins embedded in biological membranes, and they comprise specific proteins that control the flow of ion transporters in and out of cells, playing crucial roles in the biological functions of different cells. They maintain the homeostasis of water and ion metabolism by facilitating ion transport and participate in the physiological processes of neurons and glial cells by regulating signaling pathways. Neurodegenerative diseases are a group of disorders characterized by the progressive loss of neurons in the central nervous system (CNS) or peripheral nervous system (PNS). Despite significant progress in understanding the pathophysiological processes of various neurological diseases in recent years, effective treatments for mitigating the damage caused by these diseases remain inadequate. Increasing evidence suggests that ion channels are closely associated with neuroinflammation; oxidative stress; and the characteristic proteins in neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Therefore, studying the pathogenic mechanisms closely related to ion channels in neurodegenerative diseases can help identify more effective therapeutic targets for treating neurodegenerative diseases. Here, we discuss the progress of research on ion channels in different neurodegenerative diseases and emphasize the feasibility and potential of treating such diseases from the perspective of ion channels.
Collapse
Affiliation(s)
- Yuxuan Li
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.L.); (J.F.)
| | - Jingxuan Fu
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.L.); (J.F.)
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China
| | - Hui Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.L.); (J.F.)
| |
Collapse
|
26
|
El Din RH, Thabit S. Quinic acid protects against the development of Huntington's disease in Caenorhabditis elegans model. BMC Complement Med Ther 2024; 24:377. [PMID: 39468600 PMCID: PMC11514749 DOI: 10.1186/s12906-024-04670-4] [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: 07/11/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Quinic acid (QA), a cyclitol and cyclohexanecarboxylic acid, is a natural product that is present and can be isolated from edible herbals like tea, coffee and several fruits and vegetables. It was previously reported that QA exerted antioxidant and neuroprotective activity against dementia. However, it was not tested for its neuroprotective potential against Huntington's disease (HD). Since aging related disorders are greatly linked to oxidative stress conditions, we focused on testing the oxidative stress resistant activity and protective effect of QA against the development of HD by using the multicellular Caenorhabditis elegans (C. elegans) worm model. METHODS Firstly, QA was tested for its oxidative stress resistant properties. In survival assay, wild type and mutant skn-1 and daf-16 worms were exposed to oxidative stress conditions by using H2O2. Activation of SKN-1 pathway and expression of its downstream genes gcs-1 and gst-4 were also tested. Secondly, the effect of QA was evaluated on HD by testing its ability to decrease the formation of polyQ150 aggregates. Furthermore, its effect on the accumulation of polyglutamine (polyQ35 and polyQ40 aggregates) was tested. RESULTS Here we report that QA could improve the survival of C. elegans after exposure to oxidative stress caused by H2O2 while also exerting antioxidant effects through the activation of SKN-1/Nrf2 pathway. Moreover, QA could be a potential candidate to protect against HD due to its effects on decreasing the formation of polyQ150, polyQ35 and polyQ40 aggregates. CONCLUSIONS This study highlights the importance of QA as a natural compound in defending against oxidative stress and the development of neurodegenerative diseases like HD.
Collapse
Affiliation(s)
- Reem Hossam El Din
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, 11835, Egypt
| | - Sara Thabit
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo, 11835, Egypt.
| |
Collapse
|
27
|
Kim SY, Lim W. Break-up and recovery of harmony between direct and indirect pathways in the basal ganglia: Huntington's disease and treatment. Cogn Neurodyn 2024; 18:2909-2924. [PMID: 39555304 PMCID: PMC11564723 DOI: 10.1007/s11571-024-10125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/21/2024] [Accepted: 05/07/2024] [Indexed: 11/19/2024] Open
Abstract
The basal ganglia (BG) in the brain exhibit diverse functions for motor, cognition, and emotion. Such BG functions could be made via competitive harmony between the two competing pathways, direct pathway (DP) (facilitating movement) and indirect pathway (IP) (suppressing movement). As a result of break-up of harmony between DP and IP, there appear pathological states with disorder for movement, cognition, and psychiatry. In this paper, we are concerned about the Huntington's disease (HD), which is a genetic neurodegenerative disorder causing involuntary movement and severe cognitive and psychiatric symptoms. For the HD, the number of D2 SPNs ( N D 2 ) is decreased due to degenerative loss, and hence, by decreasing x D 2 (fraction of N D 2 ), we investigate break-up of harmony between DP and IP in terms of their competition degree C d , given by the ratio of strength of DP ( S DP ) to strength of IP ( S IP ) (i.e.,C d = S DP / S IP ). In the case of HD, the IP is under-active, in contrast to the case of Parkinson's disease with over-active IP, which results in increase in C d (from the normal value). Thus, hyperkinetic dyskinesia such as chorea (involuntary jerky movement) occurs. We also investigate treatment of HD, based on optogenetics and GP ablation, by increasing strength of IP, resulting in recovery of harmony between DP and IP. Finally, we study effect of loss of healthy synapses of all the BG cells on HD. Due to loss of healthy synapses, disharmony between DP and IP increases, leading to worsen symptoms of the HD. Supplementary Information The online version contains supplementary material available at 10.1007/s11571-024-10125-w.
Collapse
Affiliation(s)
- Sang-Yoon Kim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| | - Woochang Lim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| |
Collapse
|
28
|
Bhandari UR, Danish SM, Ahmad S, Ikram M, Nadaf A, Hasan N, Kesharwani P, Ahmad FJ. New opportunities for antioxidants in amelioration of neurodegenerative diseases. Mech Ageing Dev 2024; 221:111961. [PMID: 38960099 DOI: 10.1016/j.mad.2024.111961] [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: 04/02/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
This comprehensive review elucidates the critical role of antioxidants to mitigate oxidative stress, a common denominator in an array of neurodegenerative disorders. Oxidative stress-induced damage has been linked to the development of diseases such as Alzheimer's, Parkinson's, Huntington's disease and amyotrophic lateral sclerosis. This article examines a wide range of scientific literature and methodically delineates the several methods by which antioxidants exercise their neuroprotective benefits. It also explores into the complex relationship between oxidative stress and neuroinflammation, focusing on how antioxidants can alter signaling pathways and transcription factors to slow neurodegenerative processes. Key antioxidants, such as vitamins C and E, glutathione, and polyphenolic compounds, are tested for their ability to combat reactive oxygen and nitrogen species. The dual character of antioxidants, which operate as both direct free radical scavengers and regulators of cellular redox homeostasis, is investigated in terms of therapeutic potential. Furthermore, the study focuses on new antioxidant-based therapy techniques and their mechanisms including Nrf-2, PCG1α, Thioredoxin etc., which range from dietary interventions to targeted antioxidant molecules. Insights into ongoing clinical studies evaluating antioxidant therapies in neurodegenerative illnesses offer an insight into the translational potential of antioxidant research. Finally, this review summarizes our present understanding of antioxidant processes in neurodegenerative illnesses, providing important possibilities for future study and treatment development.
Collapse
Affiliation(s)
- Uttam Raj Bhandari
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Mohammad Danish
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shadaan Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Ikram
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Arif Nadaf
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Farhan J Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
29
|
Ebrahimi P, Davoudi E, Sadeghian R, Zadeh AZ, Razmi E, Heidari R, Morowvat MH, Sadeghian I. In vivo and ex vivo gene therapy for neurodegenerative diseases: a promise for disease modification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7501-7530. [PMID: 38775852 DOI: 10.1007/s00210-024-03141-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/01/2024] [Indexed: 10/04/2024]
Abstract
Neurodegenerative diseases (NDDs), including AD, PD, HD, and ALS, represent a growing public health concern linked to aging and lifestyle factors, characterized by progressive nervous system damage leading to motor and cognitive deficits. Current therapeutics offer only symptomatic management, highlighting the urgent need for disease-modifying treatments. Gene therapy has emerged as a promising approach, targeting the underlying pathology of diseases with diverse strategies including gene replacement, gene silencing, and gene editing. This innovative therapeutic approach involves introducing functional genetic material to combat disease mechanisms, potentially offering long-term efficacy and disease modification. With advancements in genomics, structural biology, and gene editing tools such as CRISPR/Cas9, gene therapy holds significant promise for addressing the root causes of NDDs. Significant progress in preclinical and clinical studies has demonstrated the potential of in vivo and ex vivo gene therapy to treat various NDDs, offering a versatile and precise approach in comparison to conventional treatments. The current review describes various gene therapy approaches employed in preclinical and clinical studies for the treatment of NDDs, including AD, PD, HD, and ALS, and addresses some of the key translational challenges in this therapeutic approach.
Collapse
Affiliation(s)
- Pouya Ebrahimi
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Davoudi
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | | | - Amin Zaki Zadeh
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Emran Razmi
- Arak University of Medical Sciences, Arak, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Morowvat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
30
|
Louçã M, El Akrouti D, Lemesle A, Louessard M, Dufour N, Baroin C, de la Fouchardière A, Cotter L, Jean-Jacques H, Redeker V, Perrier AL. Huntingtin lowering impairs the maturation and synchronized synaptic activity of human cortical neuronal networks derived from induced pluripotent stem cells. Neurobiol Dis 2024; 200:106630. [PMID: 39106928 DOI: 10.1016/j.nbd.2024.106630] [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: 06/11/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024] Open
Abstract
Despite growing descriptions of wild-type Huntingtin (wt-HTT) roles in both adult brain function and, more recently, development, several clinical trials are exploring HTT-lowering approaches that target both wt-HTT and the mutant isoform (mut-HTT) responsible for Huntington's disease (HD). This non-selective targeting is based on the autosomal dominant inheritance of HD, supporting the idea that mut-HTT exerts its harmful effects through a toxic gain-of-function or a dominant-negative mechanism. However, the precise amount of wt-HTT needed for healthy neurons in adults and during development remains unclear. In this study, we address this question by examining how wt-HTT loss affects human neuronal network formation, synaptic maturation, and homeostasis in vitro. Our findings establish a role of wt-HTT in the maturation of dendritic arborization and the acquisition of network-wide synchronized activity by human cortical neuronal networks modeled in vitro. Interestingly, the network synchronization defects only became apparent when more than two-thirds of the wt-HTT protein was depleted. Our study underscores the critical need to precisely understand wt-HTT role in neuronal health. It also emphasizes the potential risks of excessive wt-HTT loss associated with non-selective therapeutic approaches targeting both wt- and mut-HTT isoforms in HD patients.
Collapse
Affiliation(s)
- Mathilde Louçã
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Donya El Akrouti
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Aude Lemesle
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Morgane Louessard
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Noëlle Dufour
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Chloé Baroin
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Aurore de la Fouchardière
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Laurent Cotter
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Hélène Jean-Jacques
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell, 91198 Gif-sur-Yvette, France
| | - Virginie Redeker
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell, 91198 Gif-sur-Yvette, France
| | - Anselme L Perrier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
31
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
32
|
Chakraborty A, Sreenivasmurthy SG, Miller W, Huai W, Biswas T, Mandal SM, Boscá L, Krishnan B, Ghosh G, Hazra T. Fructose-2,6-bisphosphate restores DNA repair activity of PNKP and ameliorates neurodegenerative symptoms in Huntington's disease. Proc Natl Acad Sci U S A 2024; 121:e2406308121. [PMID: 39298485 PMCID: PMC11441552 DOI: 10.1073/pnas.2406308121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/05/2024] [Indexed: 09/21/2024] Open
Abstract
Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3) are the two most prevalent polyglutamine (polyQ) neurodegenerative diseases, caused by CAG (encoding glutamine) repeat expansion in the coding region of the huntingtin (HTT) and ataxin-3 (ATXN3) proteins, respectively. We have earlier reported that the activity, but not the protein level, of an essential DNA repair enzyme, polynucleotide kinase 3'-phosphatase (PNKP), is severely abrogated in both HD and SCA3 resulting in accumulation of double-strand breaks in patients' brain genome. While investigating the mechanistic basis for the loss of PNKP activity and accumulation of DNA double-strand breaks leading to neuronal death, we observed that PNKP interacts with the nuclear isoform of 6-phosphofructo-2-kinase fructose-2,6-bisphosphatase 3 (PFKFB3). Depletion of PFKFB3 markedly abrogates PNKP activity without changing its protein level. Notably, the levels of both PFKFB3 and its product fructose-2,6 bisphosphate (F2,6BP), an allosteric modulator of glycolysis, are significantly lower in the nuclear extracts of postmortem brain tissues of HD and SCA3 patients. Supplementation of F2,6BP restored PNKP activity in the nuclear extracts of patients' brain. Moreover, intracellular delivery of F2,6BP restored both the activity of PNKP and the integrity of transcribed genome in neuronal cells derived from the striatum of the HD mouse. Importantly, supplementing F2,6BP rescued the HD phenotype in Drosophila, suggesting F2,6BP to serve in vivo as a cofactor for the proper functionality of PNKP and thereby, of brain health. Our results thus provide a compelling rationale for exploring the therapeutic use of F2,6BP and structurally related compounds for treating polyQ diseases.
Collapse
Affiliation(s)
- Anirban Chakraborty
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | | | - Wyatt Miller
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Weihan Huai
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Tapan Biswas
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Santi Mohan Mandal
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols-Morreale, (Consejo Superior de Investigaciones Científicas- Universidad Autónoma de Madrid), and Centro de Investigación en Red en Enfermedades Cardiovasculares, Madrid ES-28040, Spain
| | - Balaji Krishnan
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093
| | - Tapas Hazra
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
33
|
Pradhan S, Bush K, Zhang N, Pandita RK, Tsai CL, Smith C, Pandlebury DF, Gaikwad S, Leonard F, Nie L, Tao A, Russell W, Yuan S, Choudhary S, Ramos KS, Elferink C, Wairkar YP, Tainer JA, Thompson LM, Pandita TK, Sarkar PS. Chromatin remodeler BRG1 recruits huntingtin to repair DNA double-strand breaks in neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613927. [PMID: 39345557 PMCID: PMC11429940 DOI: 10.1101/2024.09.19.613927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Persistent DNA double-strand breaks (DSBs) are enigmatically implicated in neurodegenerative diseases including Huntington's disease (HD), the inherited late-onset disorder caused by CAG repeat elongations in Huntingtin (HTT). Here we combine biochemistry, computation and molecular cell biology to unveil a mechanism whereby HTT coordinates a Transcription-Coupled Non-Homologous End-Joining (TC-NHEJ) complex. HTT joins TC-NHEJ proteins PNKP, Ku70/80, and XRCC4 with chromatin remodeler Brahma-related Gene 1 (BRG1) to resolve transcription-associated DSBs in brain. HTT recruitment to DSBs in transcriptionally active gene- rich regions is BRG1-dependent while efficient TC-NHEJ protein recruitment is HTT-dependent. Notably, mHTT compromises TC-NHEJ interactions and repair activity, promoting DSB accumulation in HD tissues. Importantly, HTT or PNKP overexpression restores TC-NHEJ in a Drosophila HD model dramatically improving genome integrity, motor defects, and lifespan. Collective results uncover HTT stimulation of DSB repair by organizing a TC-NHEJ complex that is impaired by mHTT thereby implicating dysregulation of transcription-coupled DSB repair in mHTT pathophysiology. Highlights BRG1 recruits HTT and NHEJ components to transcriptionally active DSBs.HTT joins BRG1 and PNKP to efficiently repair transcription related DSBs in brain.Mutant HTT impairs the functional integrity of TC-NHEJ complex for DSB repair.HTT expression improves DSB repair, genome integrity and phenotypes in HD flies.
Collapse
|
34
|
Ballarin G, Biasiotto M, Reisbitzer A, Hegels M, Bolte M, Krauß S, Berdnikova DV. A novel aurone RNA CAG binder inhibits the huntingtin RNA-protein interaction. RSC Med Chem 2024; 15:3092-3096. [PMID: 39309355 PMCID: PMC11411626 DOI: 10.1039/d4md00403e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/14/2024] [Indexed: 09/25/2024] Open
Abstract
Huntington's disease (HD) is a devastating, incurable condition whose pathophysiological mechanism relies on mutant RNA CAG repeat expansions. Aberrant recruitment of RNA-binding proteins by mutant CAG hairpins contributes to the progress of neurodegeneration. In this work, we identified a novel binder based on an aurone scaffold that reduces the level of binding of HTT mRNA to the MID1 protein in vitro. The obtained results introduce aurones as a novel platform for the design of functional ligands for disease-related RNA sequences.
Collapse
Affiliation(s)
- Giovanna Ballarin
- University of Padova, School of Pharmaceutical Sciences via Marzolo 5 35131 Padova Italy
- Institut für Biologie, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
- Organische Chemie II, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
| | - Maddalena Biasiotto
- University of Padova, School of Pharmaceutical Sciences via Marzolo 5 35131 Padova Italy
- Institut für Biologie, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
- Organische Chemie II, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
| | - Annika Reisbitzer
- Institut für Biologie, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
| | - Marlen Hegels
- Organische Chemie II, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
| | - Michael Bolte
- Institut für Anorganische Chemie, J.-W.-Goethe-Universität Max-von-Laue-Str. 7 60438 Frankfurt-am-Main Germany
| | - Sybille Krauß
- Institut für Biologie, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
| | - Daria V Berdnikova
- Organische Chemie II, Universität Siegen Adolf-Reichwein-Str. 2 57076 Siegen Germany
| |
Collapse
|
35
|
Xu H, Li H, Zhang P, Gao Y, Ma H, Gao T, Liu H, Hua W, Zhang L, Zhang X, Yang P, Liu J. The functions of exosomes targeting astrocytes and astrocyte-derived exosomes targeting other cell types. Neural Regen Res 2024; 19:1947-1953. [PMID: 38227520 PMCID: PMC11040311 DOI: 10.4103/1673-5374.390961] [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: 05/27/2023] [Revised: 07/20/2023] [Accepted: 09/08/2023] [Indexed: 01/17/2024] Open
Abstract
Astrocytes are the most abundant glial cells in the central nervous system; they participate in crucial biological processes, maintain brain structure, and regulate nervous system function. Exosomes are cell-derived extracellular vesicles containing various bioactive molecules including proteins, peptides, nucleotides, and lipids secreted from their cellular sources. Increasing evidence shows that exosomes participate in a communication network in the nervous system, in which astrocyte-derived exosomes play important roles. In this review, we have summarized the effects of exosomes targeting astrocytes and the astrocyte-derived exosomes targeting other cell types in the central nervous system. We also discuss the potential research directions of the exosome-based communication network in the nervous system. The exosome-based intercellular communication focused on astrocytes is of great significance to the biological and/or pathological processes in different conditions in the brain. New strategies may be developed for the diagnosis and treatment of neurological disorders by focusing on astrocytes as the central cells and utilizing exosomes as communication mediators.
Collapse
Affiliation(s)
- Hongye Xu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - He Li
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Emergency, Naval Hospital of Eastern Theater, Zhoushan, Zhejiang Province, China
| | - Ping Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuan Gao
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hongyu Ma
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tianxiang Gao
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hanchen Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Weilong Hua
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lei Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaoxi Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Pengfei Yang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianmin Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
36
|
Lisowski P, Lickfett S, Rybak-Wolf A, Menacho C, Le S, Pentimalli TM, Notopoulou S, Dykstra W, Oehler D, López-Calcerrada S, Mlody B, Otto M, Wu H, Richter Y, Roth P, Anand R, Kulka LAM, Meierhofer D, Glazar P, Legnini I, Telugu NS, Hahn T, Neuendorf N, Miller DC, Böddrich A, Polzin A, Mayatepek E, Diecke S, Olzscha H, Kirstein J, Ugalde C, Petrakis S, Cambridge S, Rajewsky N, Kühn R, Wanker EE, Priller J, Metzger JJ, Prigione A. Mutant huntingtin impairs neurodevelopment in human brain organoids through CHCHD2-mediated neurometabolic failure. Nat Commun 2024; 15:7027. [PMID: 39174523 PMCID: PMC11341898 DOI: 10.1038/s41467-024-51216-w] [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: 06/03/2023] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
Expansion of the glutamine tract (poly-Q) in the protein huntingtin (HTT) causes the neurodegenerative disorder Huntington's disease (HD). Emerging evidence suggests that mutant HTT (mHTT) disrupts brain development. To gain mechanistic insights into the neurodevelopmental impact of human mHTT, we engineered male induced pluripotent stem cells to introduce a biallelic or monoallelic mutant 70Q expansion or to remove the poly-Q tract of HTT. The introduction of a 70Q mutation caused aberrant development of cerebral organoids with loss of neural progenitor organization. The early neurodevelopmental signature of mHTT highlighted the dysregulation of the protein coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2), a transcription factor involved in mitochondrial integrated stress response. CHCHD2 repression was associated with abnormal mitochondrial morpho-dynamics that was reverted upon overexpression of CHCHD2. Removing the poly-Q tract from HTT normalized CHCHD2 levels and corrected key mitochondrial defects. Hence, mHTT-mediated disruption of human neurodevelopment is paralleled by aberrant neurometabolic programming mediated by dysregulation of CHCHD2, which could then serve as an early interventional target for HD.
Collapse
Affiliation(s)
- Pawel Lisowski
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin, Berlin, Germany
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec n/Warsaw, Poland
| | - Selene Lickfett
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Institute of Anatomy II, Heinrich-Heine-University, Düsseldorf, Germany
| | - Agnieszka Rybak-Wolf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Organoid Platform, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Carmen Menacho
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Stephanie Le
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Tancredi Massimo Pentimalli
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Charité - Universitätsmedizin, Berlin, Germany
| | - Sofia Notopoulou
- Institute of Applied Biosciences (INAB), Centre For Research and Technology Hellas (CERTH), Thessaloniki, Greece
| | - Werner Dykstra
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands
| | - Daniel Oehler
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | | | - Barbara Mlody
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Centogene, Rostock, Germany
| | - Maximilian Otto
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Haijia Wu
- Institute of Molecular Medicine, Medical School, Hamburg, Germany
| | | | - Philipp Roth
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Linda A M Kulka
- Institute of Physiological Chemistry, Martin-Luther-University, Halle-Wittenberg, Germany
| | - David Meierhofer
- Quantitative RNA Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Petar Glazar
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Quantitative RNA Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ivano Legnini
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Human Technopole, Milan, Italy
| | - Narasimha Swamy Telugu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tobias Hahn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Nancy Neuendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Duncan C Miller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Annett Böddrich
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Sebastian Diecke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Heidi Olzscha
- Institute of Molecular Medicine, Medical School, Hamburg, Germany
- Institute of Physiological Chemistry, Martin-Luther-University, Halle-Wittenberg, Germany
| | - Janine Kirstein
- Cell Biology, University of Bremen, Bremen, Germany
- Leibniz Institute on Aging - Fritz-Lipmann Institute, Jena, Germany
| | - Cristina Ugalde
- Instituto de Investigación Hospital 12 de Octubre (i + 12), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Spyros Petrakis
- Institute of Applied Biosciences (INAB), Centre For Research and Technology Hellas (CERTH), Thessaloniki, Greece
| | - Sidney Cambridge
- Institute of Anatomy II, Heinrich-Heine-University, Düsseldorf, Germany
- Dr. Senckenberg Anatomy, Anatomy II, Goethe-University, Frankfurt, Germany
| | - Nikolaus Rajewsky
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
- National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Ralf Kühn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Erich E Wanker
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy; School of Medicine and Health, Technical University of Munich and German Center for Mental Health (DZPG), Munich, Germany
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, UK
| | - Jakob J Metzger
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| | - Alessandro Prigione
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
37
|
Fanti R, Ayoubi R, Alende C, Fotouhi M, González Bolívar S, Chandrasekaran R, Southern K, Edwards AM, Harding RJ, Laflamme C. A guide to selecting high-performing antibodies for Huntingtin (UniProt ID: P42858) for use in western blot, immunoprecipitation, and immunofluorescence. F1000Res 2024; 13:922. [PMID: 39257448 PMCID: PMC11384196 DOI: 10.12688/f1000research.153670.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 09/12/2024] Open
Abstract
Huntingtin encodes a 3144 amino acid protein, with a polyglutamine repeat tract at the N-terminus. Expansion of this repeat tract above a pathogenic threshold of 36 repeats is the causative mutation of Huntington's disease, a neurodegenerative disorder characterized by loss of striatal neurons. Here we have characterized twenty Huntingtin commercial antibodies for western blot, immunoprecipitation, and immunofluorescence using a standardized experimental protocol based on comparing read-outs in knockout cell lines and isogenic parental controls. These studies are part of a larger, collaborative initiative seeking to address antibody reproducibility issues by characterizing commercially available antibodies for human proteins and publishing the results openly as a resource for the scientific community. While use of antibodies and protocols vary between laboratories, we encourage readers to use this report as a guide to select the most appropriate antibodies for their specific needs.
Collapse
Affiliation(s)
- Rebeka Fanti
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Riham Ayoubi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Charles Alende
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Maryam Fotouhi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Sara González Bolívar
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Renu Chandrasekaran
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Kathleen Southern
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Aled M. Edwards
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Rachel J. Harding
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Carl Laflamme
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - NeuroSGC/YCharOS/EDDU collaborative group
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - ABIF consortium
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Nateghi B, Keraudren R, Boulay G, Bazin M, Goupil C, Canet G, Loiselle A, St-Amour I, Planel E, Soulet D, Hébert SS. Beneficial effects of miR-132/212 deficiency in the zQ175 mouse model of Huntington's disease. Front Neurosci 2024; 18:1421680. [PMID: 39170678 PMCID: PMC11337869 DOI: 10.3389/fnins.2024.1421680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Huntington's disease (HD) is a rare genetic neurodegenerative disorder caused by an expansion of CAG repeats in the Huntingtin (HTT) gene. One hypothesis suggests that the mutant HTT gene contributes to HD neuropathology through transcriptional dysregulation involving microRNAs (miRNAs). In particular, the miR-132/212 cluster is strongly diminished in the HD brain. This study explores the effects of miR-132/212 deficiency specifically in adult HD zQ175 mice. The absence of miR-132/212 did not impact body weight, body temperature, or survival rates. Surprisingly, miR-132/212 loss seemed to alleviate, in part, the effects on endogenous Htt expression, HTT inclusions, and neuronal integrity in HD zQ175 mice. Additionally, miR-132/212 depletion led to age-dependent improvements in certain motor functions. Transcriptomic analysis revealed alterations in HD-related networks in WT- and HD zQ175-miR-132/212-deficient mice, including significant overlap in BDNF and Creb1 signaling pathways. Interestingly, however, a higher number of miR-132/212 gene targets was observed in HD zQ175 mice lacking the miR-132/212 cluster, especially in the striatum. These findings suggest a nuanced interplay between miR-132/212 expression and HD pathogenesis, providing potential insights into therapeutic interventions. Further investigation is needed to fully understand the underlying mechanisms and therapeutic potential of modulating miR-132/212 expression during HD progression.
Collapse
Affiliation(s)
- Behnaz Nateghi
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Remi Keraudren
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Gabriel Boulay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Marc Bazin
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Claudia Goupil
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Geoffrey Canet
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Andréanne Loiselle
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Isabelle St-Amour
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale-Nationale, Québec, QC, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Denis Soulet
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Sébastien S. Hébert
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
39
|
Allen S, O'Reilly D, Miller R, Sapp E, Summers A, Paquette J, Echeverria Moreno D, Bramato B, McHugh N, Yamada K, Aronin N, DiFiglia M, Khvorova A. mRNA Nuclear Clustering Leads to a Difference in Mutant Huntingtin mRNA and Protein Silencing by siRNAs In Vivo. Nucleic Acid Ther 2024; 34:164-172. [PMID: 39023561 PMCID: PMC11387003 DOI: 10.1089/nat.2024.0027] [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] [Indexed: 07/20/2024] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by CAG repeat expansion in the first exon of the huntingtin gene (HTT). Oligonucleotide therapeutics, such as short interfering RNA (siRNA), reduce levels of huntingtin mRNA and protein in vivo and are considered a viable therapeutic strategy. However, the extent to which they silence huntingtin mRNA in the nucleus is not established. We synthesized siRNA cross-reactive to mouse (wild-type) Htt and human (mutant) HTT in a divalent scaffold and delivered to two mouse models of HD. In both models, divalent siRNA sustained lowering of wild-type Htt, but not mutant HTT mRNA expression in striatum and cortex. Near-complete silencing of both mutant HTT protein and wild-type HTT protein was observed in both models. Subsequent fluorescent in situ hybridization analysis shows that divalent siRNA acts predominantly on cytoplasmic mutant HTT transcripts, leaving clustered mutant HTT transcripts in the nucleus largely intact in treated HD mouse brains. The observed differences between mRNA and protein levels, exaggerated in the case of extended repeats, might apply to other repeat-associated neurological disorders.
Collapse
Affiliation(s)
- Sarah Allen
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Daniel O'Reilly
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Rachael Miller
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Ellen Sapp
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ashley Summers
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Joseph Paquette
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Dimas Echeverria Moreno
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Brianna Bramato
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas McHugh
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Ken Yamada
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Neil Aronin
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Marian DiFiglia
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
40
|
İşcan D, Çetinkaya Y. Cardiac autonomic involvement in Huntington's disease. Neurol Sci 2024; 45:3823-3828. [PMID: 38436789 PMCID: PMC11254994 DOI: 10.1007/s10072-024-07428-5] [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: 12/12/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Huntington's disease (HD) is known as a neurodegenerative disease with movement disorder and cognitive impairment; autonomic involvement is also becoming common in some recent studies. The aim of this study is to demonstrate the presence of cardiac autonomic involvement in HD patients. METHOD Time and frequency domain parameters obtained from the 24-h Holter ECG(hECG) were compared between 20 HD patients and 20 healthy control subjects. RESULTS Fourteen HD patients had tachycardia, bradycardia, and extra beats. Interval between two heartbeats, normal-to-normal (NN), standard deviation of all normal-to-normal (SDNN), square root of the mean of the sum of the squares of the differences between consecutive N-N intervals in ms (rMSSD), and the ratio of the number of consecutive pairs of N-N intervals that differ by more than 50 ms to the total number of N-N intervals (pNN50) were all significantly higher in the patient group than in the control group during 24-h hECG monitoring. However, hECG monitoring showed that the patient group had significantly higher values of the frequency-domain metrics high frequency (HF) than the control group did (P = 0.003). Very low frequency (VLF) was lower in the patient group (P = 0.009). There was no difference in low frequency (LF) in both groups. In comparison to the control group, LF/HF was much reduced in the patient group (P = 0.001). CONCLUSION Cardiac disfunction increases, and autonomic functions change in HD, but more comprehensive studies are needed to distinguish sympathetic and parasympathetic involvement.
Collapse
Affiliation(s)
- Dilek İşcan
- Department of Neurology, Faculty of Medicine, Niğde Ömer Halisdemir University, 51240, Niğde, Turkey.
| | - Yakup Çetinkaya
- Department of Cardiology, Faculty of Medicine, Niğde Ömer Halisdemir University, 51240, Niğde, Turkey
| |
Collapse
|
41
|
Wang Y, Niu K, Shi Y, Zhou F, Li X, Li Y, Chen T, Zhang Y. A review: targeting UBR5 domains to mediate emerging roles and mechanisms - chance or necessity? Int J Surg 2024; 110:4947-4964. [PMID: 38701508 PMCID: PMC11326040 DOI: 10.1097/js9.0000000000001541] [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: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
Ubiquitinases are known to catalyze ubiquitin chains on target proteins to regulate various physiological functions like cell proliferation, autophagy, apoptosis, and cell cycle progression. As a member of E3 ligase, ubiquitin protein ligase E3 component n-recognin 5 (UBR5) belongs to the HECT E3 ligase and has been reported to be correlated with various pathophysiological processes. In this review, the authors give a comprehensive insight into the structure and function of UBR5. The authors discuss the specific domains of UBR5 and explore their biological functions separately. Furthermore, the authors describe the involvement of UBR5 in different pathophysiological conditions, including immune response, virus infection, DNA damage response, and protein quality control. Moreover, the authors provide a thorough summary of the important roles and regulatory mechanisms of UBR5 in cancers and other diseases. On the whole, investigating the domains and functions of UBR5, elucidating the underlying mechanisms of UBR5 with various substrates in detail may provide new theoretical basis for the treatment of diseases, including cancers, which could improve future studies to construct novel UBR5-targeted therapy strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| |
Collapse
|
42
|
Jambi EJ, Alamri A, Afzal M, Al-Abbasi FA, Al-Qahtani SD, Almalki NAR, Bawadood AS, Alzarea SI, Sayyed N, Kazmi I. 6-shogaol against 3-Nitropropionic acid-induced Huntington's disease in rodents: Based on molecular docking/targeting pro-inflammatory cytokines/NF-κB-BDNF-Nrf2 pathway. PLoS One 2024; 19:e0305358. [PMID: 39008492 PMCID: PMC11249262 DOI: 10.1371/journal.pone.0305358] [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: 02/17/2024] [Accepted: 05/27/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is an extremely harmful autosomal inherited neurodegenerative disease. Motor dysfunction, mental disorder, and cognitive deficits are the characteristic features of this disease. The current study examined whether 6-shogaol has a protective effect against 3-Nitropropionic Acid (3-NPA)-induced HD in rats. METHODS A total of thirty male Wistar rats received 6-shogaol (10 and 20 mg/kg, per oral) an hour before injection of 3-NPA (10 mg/kg i.p.) for 15 days. Behavioral tests were performed, including narrow beam walk, rotarod test, and grip strength test. Biochemical tests promoting oxidative stress were evaluated [superoxide dismutase (SOD), reduced glutathione (GSH), catalase (CAT) and malondialdehyde (MDA)], including changes to neurotransmitters serotonin (5-HT), dopamine (DA), norepinephrine (NE), homovanillic acid (HVA), (3,4-dihydroxyphenylacetic acid (DOPAC), γ-aminobutyric acid (GABA), and 5-hydroxy indole acetic acid (5-HIAA), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), interleukins-1β (IL-1β), IL-6, brain-derived neurotrophic factor (BDNF), and nuclear factor erythroid 2-related factor 2 (Nrf2). The 6-shogaol was docked to the active site of TNF-α (2AZ5), NF-κB (1SVC), BDNF) [1B8M], and Nrf2 [5FZN] proteins using AutoDock tools. RESULTS The 6-shogaol group significantly improved behavioral activity over the 3-NPA-injected control rats. Moreover, 3-NPA-induced significantly altered neurotransmitters, biochemical and neuroinflammatory indices, which could efficiently be reversed by 6-shogaol. The 6-shogaol showed favorable negative binding energies at -9.271 (BDNF) kcal/mol. CONCLUSIONS The present investigation demonstrated the neuroprotective effects of 6-shogaol in an experimental animal paradigm against 3-NPA-induced HD in rats. The suggested mechanism is supported by immunohistochemical analysis and western blots, although more research is necessary for definite confirmation.
Collapse
Affiliation(s)
- Ebtihaj J. Jambi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salwa D. Al-Qahtani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Naif A. R. Almalki
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Aljouf, Sakaka, Saudi Arabia
| | - Nadeem Sayyed
- School of Pharmacy, Glocal University, Saharanpur, India
| | - Imran Kazmi
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
43
|
Cordeiro LM, Soares FAA, Arantes LP. Evaluating polyglutamine protein aggregation and toxicity in transgenic Caenorhabditis elegans models of Huntington's disease. Methods Cell Biol 2024; 192:115-130. [PMID: 39863386 DOI: 10.1016/bs.mcb.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by a repeat of the cytosine-adenine-guanine trinucleotide (CAG) in the huntingtin gene (HTT). This results in the translation of a mutant huntingtin (mHTT) protein with an abnormally long polyglutamine (polyQ) repeat. The pathology of HD leads to neuronal cell loss, motor abnormalities, and dementia. Currently, the pathogenesis of HD remains incompletely understood, and available treatments only address symptoms. Caenorhabditis elegans has been used as a model for neurodegenerative diseases, enabling the exploration of the molecular, cellular, and physiological mechanisms underlying HD pathogenesis. It also facilitates the investigation of potential therapeutic targets and interventions. Here, we describe common experiments employed to assess polyQ aggregation and toxicity in transgenic C. elegans models of HD, utilizing fluorescent markers to detect protein aggregation and neuron degeneration, in addition to specific behavioral assays (thrash frequency, nose touch response, and octanol response).
Collapse
Affiliation(s)
- Larissa Marafiga Cordeiro
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Santa Maria, RS, Brazil
| | - Félix Alexandre Antunes Soares
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Santa Maria, RS, Brazil
| | | |
Collapse
|
44
|
Stanisławska-Sachadyn A, Krzemiński M, Zielonka D, Krygier M, Ziętkiewicz E, Sławek J, Limon J. Sex contribution to average age at onset of Huntington's disease depends on the number of (CAG) n repeats. Sci Rep 2024; 14:15729. [PMID: 38977715 PMCID: PMC11231309 DOI: 10.1038/s41598-024-64105-5] [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: 12/22/2023] [Accepted: 06/05/2024] [Indexed: 07/10/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by the extension of the CAG repeats in exon 1 of the HTT gene and is transmitted in a dominant manner. The present study aimed to assess whether patients' sex, in the context of mutated and normal allele length, contributes to age on onset (AO) of HD. The study population comprised a large cohort of 3723 HD patients from the European Huntington's Disease Network's REGISTRY database collected at 160 sites across 17 European countries and in one location outside Europe. The data were analyzed using regression models and factorial analysis of variance (ANOVA) considering both mutated allele length and sex as predictors of patients' AO. AO, as described by the rater's estimate, was found to be later in affected women than in men across the whole population. This difference was most pronounced in a subgroup of 1273 patients with relatively short variants of the mutated allele (40-45 CAG repeats) and normal alleles in a higher half of length distribution-namely, more than 17 CAG repeats; however, it was also observed in each group. Our results presented in this observational study point to sex-related differences in AO, most pronounced in the presence of the short mutated and long normal allele, which may add to understanding the dynamics of AO in Huntington's Disease.Trial registration: ClinicalTrials.gov identifier NCT01590589.
Collapse
Affiliation(s)
- Anna Stanisławska-Sachadyn
- Department of Biotechnology and Microbiology, Gdańsk University of Technology, 80-233, Gdańsk, Poland.
- Department of Biology and Medical Genetics, Medical University of Gdańsk, 80-211, Gdańsk, Poland.
- BioTechMed Center, Gdańsk University of Technology, Narutowicza 11/12, 80-233, Gdańsk, Poland.
| | - Michał Krzemiński
- Institute of Applied Mathematics , Gdańsk University of Technology, 80-233, Gdańsk, Poland
| | - Daniel Zielonka
- Department of Public Health, Poznań University of Medical Sciences, 60-812, Poznan, Poland
| | - Magdalena Krygier
- Department of Developmental Neurology, Medical University of Gdansk, 80-952, Gdańsk, Poland
| | - Ewa Ziętkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, 60-479, Poznan, Poland
| | - Jarosław Sławek
- Department of Neurology, St. Adalbert Hospital, Copernicus PL, 80-462,, Gdańsk, Poland
- Department of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdańsk, 80-211, Gdańsk, Poland
| | - Janusz Limon
- Department of Medical Ethics, Medical University of Gdańsk, 80-211, Gdańsk, Poland
| |
Collapse
|
45
|
Hwang YS, Jo S, Kim GH, Lee JY, Ryu HS, Oh E, Lee SH, Kim YS, Chung SJ. Clinical and Genetic Characteristics Associated With Survival Outcome in Late-Onset Huntington's Disease in South Korea. J Clin Neurol 2024; 20:394-401. [PMID: 38627228 PMCID: PMC11220345 DOI: 10.3988/jcn.2023.0329] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/29/2023] [Accepted: 11/12/2023] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND AND PURPOSE The onset of Huntington's disease (HD) usually occurs before the age of 50 years, and the median survival time from onset is 15 years. We investigated survival in patients with late-onset HD (LoHD) (age at onset ≥60 years) and the associations of the number of mutant CAG repeats and age at onset (AAO) with survival in patients with HD. METHODS Patients with genetically confirmed HD at six referral centers in South Korea between 2000 and 2020 were analyzed retrospectively. Baseline demographic, clinical, and genetic characteristics and the survival status as at December 2020 were collected. RESULTS Eighty-seven patients were included, comprising 26 with LoHD (AAO=68.77±5.91 years, mean±standard deviation; 40.54±1.53 mutant CAG repeats) and 61 with common-onset HD (CoHD) (AAO=44.12±8.61 years, 44.72±4.27 mutant CAG repeats). The ages at death were 77.78±7.46 and 53.72±10.86 years in patients with LoHD and CoHD, respectively (p<0.001). The estimated survival time was 15.21±2.49 years for all HD patients, and 10.74±1.95 and 16.15±2.82 years in patients with LoHD and CoHD, respectively. More mutant CAG repeats and higher AAO were associated with shorter survival (hazard ratio [HR]=1.05, 95% confidence interval [CI]=1.01-1.09, p=0.019; and HR=1.17, 95% CI=1.03-1.31, p=0.013; respectively) for all HD patients. The LoHD group showed no significant factors associated with survival after disease onset, whereas the number of mutant CAG repeats had a significant effect (HR=1.12, 95% CI=1.01-1.23, p=0.034) in the CoHD group. CONCLUSIONS Survival after disease onset was shorter in patients with LoHD than in those with CoHD. More mutant CAG repeats and higher AAO were associated with shorter survival in patients with HD.
Collapse
Affiliation(s)
- Yun Su Hwang
- Department of Neurology, Jeonbuk National University Medical School and Hospital, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gu-Hwan Kim
- Medical Genetic Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jee-Young Lee
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University Medical College, Seoul, Korea
| | - Ho-Sung Ryu
- Department of Neurology, Kyungpook National University Hospital, Daegu, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, Korea
| | - Seung-Hwan Lee
- Department of Neurology, Kangwon National University Hospital, Chuncheon, Korea
| | - Young Seo Kim
- Department of Neurology, Wonkwang University School of Medicine, Iksan, Korea
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
46
|
Chahla C, Kovacic H, Ferhat L, Leloup L. Pathological Impact of Redox Post-Translational Modifications. Antioxid Redox Signal 2024; 41:152-180. [PMID: 38504589 DOI: 10.1089/ars.2023.0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Oxidative stress is involved in the development of several pathologies. The different reactive oxygen species (ROS) produced during oxidative stress are at the origin of redox post-translational modifications (PTMs) on proteins and impact nucleic acids and lipids. This review provides an overview of recent data on cysteine and methionine oxidation and protein carbonylation following oxidative stress in a pathological context. Oxidation, like nitration, is a selective process and not all proteins are impacted. It depends on multiple factors, including amino acid environment, accessibility, and physical and chemical properties, as well as protein structures. Thiols can undergo reversible oxidations and others that are irreversible. On the contrary, carbonylation represents irreversible PTM. To date, hundreds of proteins were shown to be modified by ROS and reactive nitrogen species (RNS). We reviewed recent advances in the impact of redox-induced PTMs on protein functions and activity, as well as its involvement in disease development or treatment. These data show a complex situation of the involvement of redox PTM on the function of targeted proteins. Many proteins can have their activity decreased by the oxidation of cysteine thiols or methionine S-methyl thioethers, while for other proteins, this oxidation will be activating. This complexity of redox PTM regulation suggests that a global antioxidant therapeutic approach, as often proposed, is unlikely to be effective. However, the specificity of the effect obtained by targeting a cysteine or methionine residue to be able to inactivate or activate a particular protein represents a major interest if it is possible to consider this targeting from a therapeutic point of view with our current pharmacological tools. Antioxid. Redox Signal. 41, 152-180.
Collapse
Affiliation(s)
- Charbel Chahla
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| | - Hervé Kovacic
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| | - Lotfi Ferhat
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| | - Ludovic Leloup
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| |
Collapse
|
47
|
Bonavita R, Di Martino R, Cortone G, Prodomo A, Di Gennaro M, Scerra G, Panico V, Nuzzo S, Salvatore M, Williams SV, Vitale F, Caporaso MG, D’Agostino M, Pisani FM, Fleming A, Renna M. A method for the analysis of the oligomerization profile of the Huntington's disease-associated, aggregation-prone mutant huntingtin protein by isopycnic ultracentrifugation. Front Mol Biosci 2024; 11:1420691. [PMID: 38993838 PMCID: PMC11236693 DOI: 10.3389/fmolb.2024.1420691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Conformational diseases, such as Alzheimer's, Parkinson's and Huntington's diseases as well as ataxias and fronto-temporal disorders, are part of common class of neurological disorders characterised by the aggregation and progressive accumulation of mutant proteins which display aberrant conformation. In particular, Huntington's disease (HD) is caused by mutations leading to an abnormal expansion in the polyglutamine (poly-Q) tract of the huntingtin protein (HTT), leading to the formation of inclusion bodies in neurons of affected patients. Furthermore, recent experimental evidence is challenging the conventional view of the disease by revealing the ability of mutant HTT to be transferred between cells by means of extracellular vesicles (EVs), allowing the mutant protein to seed oligomers involving both the mutant and wild type forms of the protein. There is still no successful strategy to treat HD. In addition, the current understanding of the biological processes leading to the oligomerization and aggregation of proteins bearing the poly-Q tract has been derived from studies conducted on isolated poly-Q monomers and oligomers, whose structural properties are still unclear and often inconsistent. Here we describe a standardised biochemical approach to analyse by isopycnic ultracentrifugation the oligomerization of the N-terminal fragment of mutant HTT. The dynamic range of our method allows one to detect large and heterogeneous HTT complexes. Hence, it could be harnessed for the identification of novel molecular determinants responsible for the aggregation and the prion-like spreading properties of HTT in the context of HD. Equally, it provides a tool to test novel small molecules or bioactive compounds designed to inhibit the aggregation of mutant HTT.
Collapse
Affiliation(s)
- Raffaella Bonavita
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Rosaria Di Martino
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
- Institute for Endocrinology and Experimental Oncology “G. Salvatore”, National Research Council, Naples, Italy
| | - Giuseppe Cortone
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Antonello Prodomo
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Mariagrazia Di Gennaro
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
- Institute for Endocrinology and Experimental Oncology “G. Salvatore”, National Research Council, Naples, Italy
| | - Gianluca Scerra
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Valentino Panico
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | | | | | - Sarah V. Williams
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Fulvia Vitale
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Maria Gabriella Caporaso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Massimo D’Agostino
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Francesca M. Pisani
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Angeleen Fleming
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Maurizio Renna
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
48
|
Liu M, Zhao J, Xue C, Yang J, Ying L. Uncovering the ferroptosis related mechanism of laduviglusib in the cell-type-specific targets of the striatum in Huntington's disease. BMC Genomics 2024; 25:633. [PMID: 38918688 PMCID: PMC11197352 DOI: 10.1186/s12864-024-10534-5] [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] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder featured by abnormal movements, arising from the extensive neuronal loss and glial dysfunction in the striatum. Although the causes and pathogenetic mechanisms of HD are well established, the development of disease-modifying pharmacological therapies for HD remains a formidable challenge. Laduviglusib has demonstrated neuroprotective effects through the enhancement of mitochondrial function in the striatum of HD animal models. Ferroptosis is a nonapoptotic form of cell death that occurs as a consequence of lethal iron-dependent lipid peroxidation and mitochondrial dysfunction. However, the ferroptosis-related mechanisms underlying the neuroprotective effects of laduviglusib in the striatum of HD patients remain largely uncharted. In this study, we leveraged single-nucleus RNA sequencing data obtained from the striatum of HD patients in stages 2-4 to identify differentially expressed genes within distinct cell-type. We subsequently integrated these differentially expressed genes of HD, laduviglusib target genes and ferroptosis-related genes to predict the ferroptosis-related mechanisms underpinning the neuroprotective effects of laduviglusib in HD patients. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses unveiled that the effects of laduviglusib on direct pathway striatal projection neurons (dSPNs) is mainly associated with Th17 cell differentiation pathways. Conversely, its impact on indirect pathway striatal projection neurons (iSPNs) extends to the Neurotrophin signaling pathway, FoxO signaling pathway, and reactive oxygen species pathway. In microglia, laduviglusib appears to contribute to HD pathology via mechanisms related to Th17 cell differentiation and the FoxO signaling pathway. Further, molecular docking results indicated favorable binding of laduviglusib with PARP1 (associated with dSPNs and iSPNs), SCD (associated with astrocytes), ALOX5 (associated with microglia), and HIF1A (associated with dSPNs, iSPNs, and microglia). In addition, the KEGG results suggest that laduviglusib may enhance mitochondrial function and protect against neuronal loss by targeting ferroptosis-related signaling pathways, particularly mediated by ALOX5 in microglia. These findings provide valuable insights into the potential mechanisms through which laduviglusib exerts its effects on distinct cell-types within the HD striatum.
Collapse
Affiliation(s)
- Mei Liu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jinlan Zhao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, School of Pharmaceutical Science, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chengcheng Xue
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun Yang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Li Ying
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
49
|
Torricella F, Tugarinov V, Clore GM. Effects of Macromolecular Cosolutes on the Kinetics of Huntingtin Aggregation Monitored by NMR Spectroscopy. J Phys Chem Lett 2024; 15:6375-6382. [PMID: 38857530 PMCID: PMC11345868 DOI: 10.1021/acs.jpclett.4c01410] [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] [Indexed: 06/12/2024]
Abstract
The effects of two macromolecular cosolutes, specifically the polysaccharide dextran-20 and the protein lysozyme, on the aggregation kinetics of a pathogenic huntingtin exon-1 protein (hhtex1) with a 35 polyglutamine repeat, httex1Q35, are described. A unified kinetic model that establishes a direct connection between reversible tetramerization occurring on the microsecond time scale and irreversible fibril formation on a time scale of hours/days forms the basis for quantitative analysis of httex1Q35 aggregation, monitored by measuring cross-peak intensities in a series of 2D 1H-15N NMR correlation spectra acquired during the course of aggregation. The primary effects of the two cosolutes are associated with shifts in the prenucleation tetramerization equilibrium resulting in substantial changes in concentration of "preformed" httex1Q35 tetramers. Similar effects of the two cosolutes on the tetramerization equilibrium observed for a shorter, nonaggregating huntingtin variant with a 7-glutamine repeat, httex1Q7, lend confidence to the conclusions drawn from the fits to the httex1Q35 aggregation kinetics.
Collapse
Affiliation(s)
- Francesco Torricella
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0520, United States
| | - Vitali Tugarinov
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0520, United States
| | - G Marius Clore
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0520, United States
| |
Collapse
|
50
|
Niazi SK, Mariam Z, Magoola M. Engineered Antibodies to Improve Efficacy against Neurodegenerative Disorders. Int J Mol Sci 2024; 25:6683. [PMID: 38928395 PMCID: PMC11203520 DOI: 10.3390/ijms25126683] [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: 05/11/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antibodies that can selectively remove rogue proteins in the brain are an obvious choice to treat neurodegenerative disorders (NDs), but after decades of efforts, only two antibodies to treat Alzheimer's disease are approved, dozens are in the testing phase, and one was withdrawn, and the other halted, likely due to efficacy issues. However, these outcomes should have been evident since these antibodies cannot enter the brain sufficiently due to the blood-brain barrier (BBB) protectant. However, all products can be rejuvenated by binding them with transferrin, preferably as smaller fragments. This model can be tested quickly and at a low cost and should be applied to bapineuzumab, solanezumab, crenezumab, gantenerumab, aducanumab, lecanemab, donanemab, cinpanemab, and gantenerumab, and their fragments. This paper demonstrates that conjugating with transferrin does not alter the binding to brain proteins such as amyloid-β (Aβ) and α-synuclein. We also present a selection of conjugate designs that will allow cleavage upon entering the brain to prevent their exocytosis while keeping the fragments connected to enable optimal binding to proteins. The identified products can be readily tested and returned to patients with the lowest regulatory cost and delays. These engineered antibodies can be manufactured by recombinant engineering, preferably by mRNA technology, as a more affordable solution to meet the dire need to treat neurodegenerative disorders effectively.
Collapse
Affiliation(s)
| | - Zamara Mariam
- Centre for Health and Life Sciences, Coventry University, Coventry City CV1 5FB, UK;
| | | |
Collapse
|