1
|
Aborode AT, Abass OA, Nasiru S, Eigbobo MU, Nefishatu S, Idowu A, Tiamiyu Z, Awaji AA, Idowu N, Busayo BR, Mehmood Q, Onifade IA, Fakorede S, Akintola AA. RNA binding proteins (RBPs) on genetic stability and diseases. Glob Med Genet 2025; 12:100032. [PMID: 39925443 PMCID: PMC11803229 DOI: 10.1016/j.gmg.2024.100032] [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: 10/29/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 02/11/2025] Open
Abstract
RNA-binding proteins (RBPs) are integral components of cellular machinery, playing crucial roles in the regulation of gene expression and maintaining genetic stability. Their interactions with RNA molecules govern critical processes such as mRNA splicing, stability, localization, and translation, which are essential for proper cellular function. These proteins interact with RNA molecules and other proteins to form ribonucleoprotein complexes (RNPs), hence controlling the fate of target RNAs. The interaction occurs via RNA recognition motif, the zinc finger domain, the KH domain and the double stranded RNA binding motif (all known as RNA-binding domains (RBDs). These domains are found within the coding sequences (intron and exon domains), 5' untranslated regions (5'UTR) and 3' untranslated regions (3'UTR). Dysregulation of RBPs can lead to genomic instability, contributing to various pathologies, including cancer neurodegenerative diseases, and metabolic disorders. This study comprehensively explores the multifaceted roles of RBPs in genetic stability, highlighting their involvement in maintaining genomic integrity through modulation of RNA processing and their implications in cellular signalling pathways. Furthermore, it discusses how aberrant RBP function can precipitate genetic instability and disease progression, emphasizing the therapeutic potential of targeting RBPs in restoring cellular homeostasis. Through an analysis of current literature, this study aims to delineate the critical role of RBPs in ensuring genetic stability and their promise as targets for innovative therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Shaibu Nasiru
- Department of Research and Development, Healthy Africans Platform, Ibadan, Nigeria
- Department of Biochemistry, Ambrose Alli University Ekpoma, Nigeria
| | | | - Sumana Nefishatu
- Department of Biochemistry, Ambrose Alli University Ekpoma, Nigeria
| | - Abdullahi Idowu
- Department of Biological Sciences, Purdue University Fort Wayne, USA
| | - Zainab Tiamiyu
- Department of Biochemistry and Cancer Biology, Medical College of Georgia, Augusta University, USA
| | - Aeshah A. Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Nike Idowu
- Department of Chemistry, University of Nebraska-Lincoln, USA
| | | | - Qasim Mehmood
- Shifa Clinical Research Center, Shifa International Hospital, Islamabad, Pakistan
| | - Isreal Ayobami Onifade
- Department of Division of Family Health, Health Research Incorporated, New York State Department of Health, USA
| | - Sodiq Fakorede
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ashraf Akintayo Akintola
- Department of Biology Education, Teachers College & Institute for Phylogenomics and Evolution, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
2
|
van Veenhuijzen K, Tan HH, Nitert AD, van Es MA, Veldink JH, van den Berg LH, Westeneng H. Longitudinal Magnetic Resonance Imaging in Asymptomatic C9orf72 Mutation Carriers Distinguishes Phenoconverters to Amyotrophic Lateral Sclerosis or Amyotrophic Lateral Sclerosis With Frontotemporal Dementia. Ann Neurol 2025; 97:281-295. [PMID: 39487710 PMCID: PMC11740280 DOI: 10.1002/ana.27116] [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: 03/15/2024] [Revised: 10/01/2024] [Accepted: 10/05/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVE We prospectively studied asymptomatic C9orf72 mutation carriers, identifying those developing amyotrophic lateral sclerosis (ALS) or frontotemporal dementia (FTD). METHODS We enrolled 56 asymptomatic family members (AFM) with a C9orf72 mutation (AFM C9+), 132 non-carriers (AFM C9-), and 359 population-based controls. Using 3 T magnetic resonance imaging, we measured cortical thickness, gyrification, and subcortical volumes longitudinally. Linear mixed-effects models on non-converting AFM C9+ scans (n = 107) created a reference for these measurements, establishing individual atrophy patterns. Atrophy patterns from presymptomatic phenoconverters (n = 10 scans) served as a template for group comparisons and similarity assessments. Similarity with phenoconverters was quantified using Dice similarity coefficient (DSC) for cortical and Kullback-Leibler similarity (KLS) for subcortical measures. Using longitudinal similarity assessments, we predicted when participants would reach the average similarity level of phenoconverters at their first post-onset scan. RESULTS Five AFM C9+ converted to ALS or ALS-FTD. Up to 6 years before symptoms, these phenoconverters exhibited significant atrophy in frontal, temporal, parietal, and cingulate cortex, along with smaller thalamus, hippocampus, and amygdala compared to other AFM C9+. Some non-converted AFM C9+ had high DSC and KLS, approaching values of phenoconverters, whereas others, along with AFM C9- and controls, had lower values. At age 80, we predicted 27.9% (95% confidence interval, 13.2-40.1%) of AFM C9+ and no AFM C9- would reach the same DSC as phenoconverters. INTERPRETATION Distinctive atrophy patterns are visible years before symptom onset on presymptomatic scans of phenoconverters. Combining baseline and follow-up similarity measures may serve as a promising imaging biomarker for identifying those at risk of ALS or ALS-FTD. ANN NEUROL 2025;97:281-295.
Collapse
Affiliation(s)
- Kevin van Veenhuijzen
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Harold H.G. Tan
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Abram D. Nitert
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Michael A. van Es
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Jan H. Veldink
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Leonard H. van den Berg
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Henk‐Jan Westeneng
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
3
|
van den Berg LH, Rothstein JD, Shaw PJ, Babu S, Benatar M, Bucelli RC, Genge A, Glass JD, Hardiman O, Libri V, Mobach T, Oskarsson B, Pattee GL, Ravits J, Shaw CE, Weber M, Zinman L, Jafar-Nejad P, Rigo F, Lin L, Ferguson TA, Gotter AL, Graham D, Monine M, Inra J, Sinks S, Eraly S, Garafalo S, Fradette S. Safety, tolerability, and pharmacokinetics of antisense oligonucleotide BIIB078 in adults with C9orf72-associated amyotrophic lateral sclerosis: a phase 1, randomised, double blinded, placebo-controlled, multiple ascending dose study. Lancet Neurol 2024; 23:901-912. [PMID: 39059407 DOI: 10.1016/s1474-4422(24)00216-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Hexanucleotide repeat expansion of C9orf72 is a common genetic cause of amyotrophic lateral sclerosis (ALS). No C9orf72-targeted treatments are available. BIIB078 is an investigational antisense oligonucleotide targeting C9orf72 sense RNA. We aimed to assess the safety, tolerability, and pharmacokinetics of BIIB078 in participants with C9orf72-associated ALS. METHODS This phase 1, randomised controlled trial was done at 22 sites in six countries (Canada, Ireland, Netherlands, Switzerland, UK, and USA). Adults with ALS and a pathogenic repeat expansion in C9orf72 were randomly assigned within six cohorts, via Interactive Response Technology in a 3:1 ratio per cohort, to receive BIIB078 (5 mg, 10 mg, 20 mg, 35 mg, 60 mg, or 90 mg in cohorts 1-6, respectively) or placebo, via an intrathecal bolus injection. The treatment period consisted of three loading doses of study treatment, administered approximately once every 2 weeks, followed by monthly maintenance doses during a treatment period of about 3 months for cohorts 1-3 and about 6 months for cohorts 4-6. Patients and investigators were masked to treatment assignment. The primary endpoint was the incidence of adverse events and serious adverse events. This trial was registered with ClinicalTrials.gov (NCT03626012) and is completed. FINDINGS Between Sept 10, 2018, and Nov 17, 2021, 124 patients were screened for inclusion in the study. 18 patients were excluded and 106 participants were enrolled and randomly assigned to receive 5 mg (n=6), 10 mg (n=9), 20 mg (n=9), 35 mg (n=19), 60 mg (n=18), or 90 mg (n=18) of BIIB078, or placebo (n=27). 58 (55%) of 106 patients were female. All patients received at least one dose of study treatment and were included in all analyses. All participants had at least one adverse event; most adverse events were mild or moderate in severity and did not lead to treatment discontinuation. The most common adverse events in BIIB078-treated participants were falls, procedural pain, headache, and post lumbar puncture syndrome. 14 (18%) of 79 patients who received any dose of BIIB078 reported serious adverse events, compared with nine (33%) of 27 patients who received placebo. Five participants who received BIIB078 and three participants who received placebo had fatal adverse events: respiratory failure in a participant who received 10 mg BIIB078, ALS worsening in two participants who received 35 mg BIIB078, traumatic intracerebral haemorrhage in one participant who received 35 mg BIIB078, pulmonary embolism in one participant who received 60 mg BIIB078, and respiratory failure in three participants who received placebo. All deaths were assessed as not related to the study treatment by the reporting investigator. INTERPRETATION On the basis of these phase 1 study results, including secondary and exploratory findings showing no reduction in neurofilament levels and no benefit on clinical outcomes relative to the placebo cohort, BIIB078 clinical development has been discontinued. However, these results will be informative in furthering our understanding of the complex pathobiology of C9orf72-associated ALS. FUNDING Biogen.
Collapse
Affiliation(s)
| | | | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK; National Institute for Health and Care Research Sheffield Biomedical Research Centre and Clinical Research Facility, University of Sheffield and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Suma Babu
- Sean M Healey and AMG Center for ALS, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert C Bucelli
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Angela Genge
- Montreal Neurological Institute and Hospital, Montreal, Canada
| | | | - Orla Hardiman
- School of Medicine, Trinity College, Dublin, Ireland
| | - Vincenzo Libri
- University College London Institute of Neurology and National Institute for Health and Care Research (NIHR) University College London Hospitals (UCLH) Clinical Research Facility and Biomedical Research Centre, London, UK
| | - Theodore Mobach
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gary L Pattee
- Bryan Physicians Network, Lincoln, NE, USA; Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - John Ravits
- Department of Neurosciences, University of California San Diego Health, San Diego, CA, USA
| | | | - Markus Weber
- Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital St Gallen, Switzerland
| | - Lorne Zinman
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Hsiao-Nakamoto J, Chiu CL, VandeVrede L, Ravi R, Vandenberg B, De Groot J, Tsogtbaatar B, Fang M, Auger P, Gould NS, Marchioni F, Powers CA, Davis SS, Suh JH, Alkabsh J, Heuer HW, Lago AL, Scearce-Levie K, Seeley WW, Boeve BF, Rosen HJ, Berger A, Tsai R, Di Paolo G, Boxer AL, Bhalla A, Huang F. Alterations in Lysosomal, Glial and Neurodegenerative Biomarkers in Patients with Sporadic and Genetic Forms of Frontotemporal Dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579529. [PMID: 38405775 PMCID: PMC10888909 DOI: 10.1101/2024.02.09.579529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Background Frontotemporal dementia (FTD) is the most common cause of early-onset dementia with 10-20% of cases caused by mutations in one of three genes: GRN, C9orf72, or MAPT. To effectively develop therapeutics for FTD, the identification and characterization of biomarkers to understand disease pathogenesis and evaluate the impact of specific therapeutic strategies on the target biology as well as the underlying disease pathology are essential. Moreover, tracking the longitudinal changes of these biomarkers throughout disease progression is crucial to discern their correlation with clinical manifestations for potential prognostic usage. Methods We conducted a comprehensive investigation of biomarkers indicative of lysosomal biology, glial cell activation, synaptic and neuronal health in cerebrospinal fluid (CSF) and plasma from non-carrier controls, sporadic FTD (symptomatic non-carriers) and symptomatic carriers of mutations in GRN, C9orf72, or MAPT, as well as asymptomatic GRN mutation carriers. We also assessed the longitudinal changes of biomarkers in GRN mutation carriers. Furthermore, we examined biomarker levels in disease impacted brain regions including middle temporal gyrus (MTG) and superior frontal gyrus (SFG) and disease-unaffected inferior occipital gyrus (IOG) from sporadic FTD and symptomatic GRN carriers. Results We confirmed glucosylsphingosine (GlcSph), a lysosomal biomarker regulated by progranulin, was elevated in the plasma from GRN mutation carriers, both symptomatic and asymptomatic. GlcSph and other lysosomal biomarkers such as ganglioside GM2 and globoside GB3 were increased in the disease affected SFG and MTG regions from sporadic FTD and symptomatic GRN mutation carriers, but not in the IOG, compared to the same brain regions from controls. The glial biomarkers GFAP in plasma and YKL40 in CSF were elevated in asymptomatic GRN carriers, and all symptomatic groups, except the symptomatic C9orf72 mutation group. YKL40 was also increased in SFG and MTG regions from sporadic FTD and symptomatic GRN mutation carriers. Neuronal injury and degeneration biomarkers NfL in CSF and plasma, and UCHL1 in CSF were elevated in patients with all forms of FTD. Synaptic biomarkers NPTXR, NPTX1/2, and VGF were reduced in CSF from patients with all forms of FTD, with the most pronounced reductions observed in symptomatic MAPT mutation carriers. Furthermore, we demonstrated plasma NfL was significantly positively correlated with disease severity as measured by CDR+NACC FTLD SB in genetic forms of FTD and CSF NPTXR was significantly negatively correlated with CDR+NACC FTLD SB in symptomatic GRN and MAPT mutation carriers. Conclusions In conclusion, our comprehensive investigation replicated alterations in biofluid biomarkers indicative of lysosomal function, glial activation, synaptic and neuronal health across sporadic and genetic forms of FTD and unveiled novel insights into the dysregulation of these biomarkers within brain tissues from patients with GRN mutations. The observed correlations between biomarkers and disease severity open promising avenues for prognostic applications and for indicators of drug efficacy in clinical trials. Our data also implicated a complicated relationship between biofluid and tissue biomarker changes and future investigations should delve into the mechanistic underpinnings of these biomarkers, which will serve as a foundation for the development of targeted therapeutics for FTD.
Collapse
Affiliation(s)
- Jennifer Hsiao-Nakamoto
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| | - Chi-Lu Chiu
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| | - Lawren VandeVrede
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Ritesh Ravi
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Brittany Vandenberg
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Brittany Vandenberg, Washington State University, Pullman, WA 99164, USA
| | - Jack De Groot
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Jack DeGroot: Prime Medicine Inc., Cambridge, MA 02139, USA
| | | | - Meng Fang
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Paul Auger
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Paul Auger: Nurix Therapeutics, San Francisco, CA 94158, USA
| | - Neal S Gould
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Filippo Marchioni
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Casey A Powers
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Casey A. Powers: Stanford University, Stanford, CA 94305, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Jung H Suh
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Jamal Alkabsh
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Hilary W Heuer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Argentina Lario Lago
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Kimberly Scearce-Levie
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Kimberly Scearce-Levie: Cajal Neuroscience, Seattle, WA 98109, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Amy Berger
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Richard Tsai
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Gilbert Di Paolo
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
- These authors contributed equally
| | - Akhil Bhalla
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| | - Fen Huang
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| |
Collapse
|
5
|
mRNA Metabolism in Health and Disease. Biomedicines 2022; 10:biomedicines10092262. [PMID: 36140363 PMCID: PMC9496247 DOI: 10.3390/biomedicines10092262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/06/2022] [Indexed: 12/04/2022] Open
|
6
|
Cruchaga C. Integrating functional genomics with genetics to understand the biology of ALS and FTD. MED 2022; 3:226-227. [PMID: 35590152 DOI: 10.1016/j.medj.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Genetic variants in chromosome 19 are strongly associated with frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Ma et al.1 and Brown et al.2 demonstrated that this association was driven by variation in UNC13A and that the underlying mechanism involved aberrant TDP-43 localization, a known pathological hallmark of both diseases.
Collapse
Affiliation(s)
- Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA; The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Ozcan KA, Ghaffari LT, Haeusler AR. The effects of molecular crowding and CpG hypermethylation on DNA G-quadruplexes formed by the C9orf72 nucleotide repeat expansion. Sci Rep 2021; 11:23213. [PMID: 34853325 PMCID: PMC8636472 DOI: 10.1038/s41598-021-02041-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/09/2021] [Indexed: 01/09/2023] Open
Abstract
A nucleotide repeat expansion (NRE), (G4C2)n, located in a classically noncoding region of C9orf72 (C9), is the most common genetic mutation associated with ALS/FTD. There is increasing evidence that nucleic acid structures formed by the C9-NRE may both contribute to ALS/FTD, and serve as therapeutic targets, but there is limited characterization of these nucleic acid structures under physiologically and disease relevant conditions. Here we show in vitro that the C9-NRE DNA can form both parallel and antiparallel DNA G-quadruplex (GQ) topological structures and that the structural preference of these DNA GQs can be dependent on the molecular crowding conditions. Additionally, 5-methylcytosine DNA hypermethylation, which is observed in the C9-NRE locus in some patients, has minimal effects on GQ topological preferences. Finally, molecular dynamic simulations of methylated and nonmethylated GQ structures support in vitro data showing that DNA GQ structures formed by the C9-NRE DNA are stable, with structural fluctuations limited to the cytosine-containing loop regions. These findings provide new insight into the structural polymorphic preferences and stability of DNA GQs formed by the C9-NRE in both the methylated and nonmethylated states, as well as reveal important features to guide the development of upstream therapeutic approaches to potentially attenuate C9-NRE-linked diseases.
Collapse
Affiliation(s)
- Kadir A Ozcan
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Layla T Ghaffari
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aaron R Haeusler
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA. .,Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, 900 Walnut Street, JHN suite 410, Philadelphia, PA, 19107, USA.
| |
Collapse
|
8
|
Parkin beyond Parkinson’s Disease—A Functional Meaning of Parkin Downregulation in TDP-43 Proteinopathies. Cells 2021; 10:cells10123389. [PMID: 34943897 PMCID: PMC8699658 DOI: 10.3390/cells10123389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Parkin and PINK1 are key regulators of mitophagy, an autophagic pathway for selective elimination of dysfunctional mitochondria. To this date, parkin depletion has been associated with recessive early onset Parkinson’s disease (PD) caused by loss-of-function mutations in the PARK2 gene, while, in sporadic PD, the activity and abundance of this protein can be compromised by stress-related modifications. Intriguingly, research in recent years has shown that parkin depletion is not limited to PD but is also observed in other neurodegenerative diseases—especially those characterized by TDP-43 proteinopathies, such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Here, we discuss the evidence of parkin downregulation in these disease phenotypes, its emerging connections with TDP-43, and its possible functional implications.
Collapse
|