1
|
Kärkkäinen V, Saari T, Hannonen S, Rusanen M, Lehtola JM, Uusitalo H, Leinonen V, Thiede B, Kaarniranta K, Koivisto AM, Utheim TP. Altered tear fluid protein expression in persons with mild Alzheimer's disease in proteins involved in oxidative stress, protein synthesis, and energy metabolism. J Alzheimers Dis 2025:13872877251326868. [PMID: 40183343 DOI: 10.1177/13872877251326868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
BackgroundTear fluid (TF) is a protein-rich solution that reflects pathophysiological changes in Alzheimer's disease (AD).ObjectiveIn this study, we examined whether TF proteins were differently expressed in persons with mild AD dementia compared to cognitively healthy controls (CO).MethodsWe analyzed data from 53 study participants including 34 CO (mean age, 71 years; Mini-Mental State Examination [MMSE] score, 28.9 ± 1.4), and 19 patients with AD (Clinical Dementia Rating, 0.5-1; mean age, 72 years; MMSE score, 23.8 ± 2.8). All participants underwent cognitive testing, as well as neurological and ophthalmological examinations. TF was collected using Schirmer strips, and TF protein content was evaluated using mass spectrometry-based proteomics and label-free quantification.ResultsWe found that 16 proteins exhibited significantly upregulated expression in the AD group compared to the CO group (p ≤ 0.05). These proteins were NP1L4, BBOX1, CYTC, RNAS4, PCD, RNT2, AL1A3, SYSC, TPIS, CLH1, PGAM1, EIF3L, 5NTC, HNRNPA2B1, PYGL, and ERO1α. No proteins were significantly downregulated in the AD group compared to the CO group.ConclusionsOur results support the hypothesis that TF is a potential source of biomarkers for AD. Part of those proteins with altered expression have previously linked to increased oxidative stress, changed protein synthesis, and disturbed regulation of energy metabolism related to AD or neurodegenerative disease. The present results indicate the value of continued investigation of TF proteins in AD.
Collapse
Affiliation(s)
- Virve Kärkkäinen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Toni Saari
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanna Hannonen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Minna Rusanen
- Ceriatric Center, Wellbeing Services Country of North Karelia, Joensuu, Finland
| | - Juha-Matti Lehtola
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Hannu Uusitalo
- Eye and Vision Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ville Leinonen
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Anne M Koivisto
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Department of Geriatrics, Helsinki University Hospital and Department of Neurosciences, University of Helsinki, Helsinki, Finland
| | - Tor P Utheim
- Department of Ophthalmology, University of Oslo, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Xia W, Liu Y, Lu J, Cheung HH, Meng Q, Huang B. RNA methylation in neurodevelopment and related diseases. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1723-1732. [PMID: 39344412 PMCID: PMC11693867 DOI: 10.3724/abbs.2024159] [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: 02/23/2024] [Accepted: 05/23/2024] [Indexed: 10/01/2024] Open
Abstract
Biological development and genetic information transfer are governed by genetic, epigenetic, transcriptional, and posttranscriptional mechanisms. RNA methylation, the attachment of methyl (-CH 3) groups to RNA molecules, is a posttranscriptional modification that has gained increasing attention in recent years because of its role in RNA epitranscriptomics. RNA modifications (RMs) influence various aspects of RNA metabolism and are involved in the regulation of diverse biological processes and diseases. Neural cell types emerge at specific stages of brain development, and recent studies have revealed that neurodevelopment, aging, and disease are tightly linked to transcriptome dysregulation. In this review, we discuss the roles of N6-methyladenine (m6A) and 5-methylcytidine (m5C) RNA modifications in neurodevelopment, physiological functions, and related diseases.
Collapse
Affiliation(s)
- Wenjuan Xia
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou)Suzhou Affiliated Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215002China
| | - Yue Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou)Suzhou Affiliated Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215002China
| | - Jiafeng Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou)Suzhou Affiliated Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215002China
| | - Hoi-Hung Cheung
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong 999077China
| | - Qingxia Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou)Suzhou Affiliated Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215002China
| | - Boxian Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou)Suzhou Affiliated Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215002China
| |
Collapse
|
3
|
German‐Castelan L, Shanks HRC, Gros R, Saito T, Saido TC, Saksida LM, Bussey TJ, Prado MAM, Schmitz TW, Prado VF. Sex-dependent cholinergic effects on amyloid pathology: A translational study. Alzheimers Dement 2024; 20:995-1012. [PMID: 37846816 PMCID: PMC10916951 DOI: 10.1002/alz.13481] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION About two-thirds of Alzheimer's Disease (AD) patients are women, who exhibit more severe pathology and cognitive decline than men. Whether biological sex causally modulates the relationship between cholinergic signaling and amyloid pathology remains unknown. METHODS We quantified amyloid beta (Aβ) in male and female App-mutant mice with either decreased or increased cholinergic tone and examined the impact of ovariectomy and estradiol replacement in this relationship. We also investigated longitudinal changes in basal forebrain (cholinergic function) and Aβ in elderly individuals. RESULTS We show a causal relationship between cholinergic tone and amyloid pathology in males and ovariectomized female mice, which is decoupled in ovary-intact and ovariectomized females receiving estradiol. In elderly humans, cholinergic loss exacerbates Aβ. DISCUSSION Our findings emphasize the importance of reflecting human menopause in mouse models. They also support a role for therapies targeting estradiol and cholinergic signaling to reduce Aβ. HIGHLIGHTS Cholinergic tone regulates amyloid beta (Aβ) pathology in males and ovariectomized female mice. Estradiol uncouples the relationship between cholinergic tone and Aβ. In elderly humans, cholinergic loss correlates with increased Aβ in both sexes.
Collapse
Affiliation(s)
- Liliana German‐Castelan
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Hayley R. C. Shanks
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Robert Gros
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of MedicineSchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Takashi Saito
- Department of Neurocognitive ScienceInstitute of Brain ScienceNagoya City University Graduate School of Medical SciencesNagoyaJapan
- Laboratory for Proteolytic NeuroscienceRIKEN Center for Brain ScienceWako, SaitamaJapan
| | - Takaomi C. Saido
- Laboratory for Proteolytic NeuroscienceRIKEN Center for Brain ScienceWako, SaitamaJapan
| | - Lisa M. Saksida
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
| | - Timothy J. Bussey
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
| | - Marco A. M. Prado
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Taylor W. Schmitz
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's HospitalLondonOntarioCanada
| | - Vania F. Prado
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | | |
Collapse
|
4
|
Nikom D, Zheng S. Alternative splicing in neurodegenerative disease and the promise of RNA therapies. Nat Rev Neurosci 2023; 24:457-473. [PMID: 37336982 DOI: 10.1038/s41583-023-00717-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 06/21/2023]
Abstract
Alternative splicing generates a myriad of RNA products and protein isoforms of different functions from a single gene. Dysregulated alternative splicing has emerged as a new mechanism broadly implicated in the pathogenesis of neurodegenerative diseases such as Alzheimer disease, amyotrophic lateral sclerosis, frontotemporal dementia, Parkinson disease and repeat expansion diseases. Understanding the mechanisms and functional outcomes of abnormal splicing in neurological disorders is vital in developing effective therapies to treat mis-splicing pathology. In this Review, we discuss emerging research and evidence of the roles of alternative splicing defects in major neurodegenerative diseases and summarize the latest advances in RNA-based therapeutic strategies to target these disorders.
Collapse
Affiliation(s)
- David Nikom
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, USA
- Center for RNA Biology and Medicine, University of California, Riverside, Riverside, CA, USA
| | - Sika Zheng
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, USA.
- Center for RNA Biology and Medicine, University of California, Riverside, Riverside, CA, USA.
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, USA.
| |
Collapse
|
5
|
Jiang L, Li X, Wang S, Yuan Z, Cheng J. The role and regulatory mechanism of m6A methylation in the nervous system. Front Genet 2022; 13:962774. [PMID: 36118889 PMCID: PMC9474891 DOI: 10.3389/fgene.2022.962774] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenosine (m6A) modification regulates RNA translation, splicing, transport, localization, and stability at the post-transcriptional level. The m6A modification has been reported to have a wide range of effects on the nervous system, including neurogenesis, cerebellar development, learning, cognition, and memory, as well as the occurrence and development of neurological disorders. In this review, we aim to summarize the findings on the role and regulatory mechanism of m6A modification in the nervous system, to reveal the molecular mechanisms of neurodevelopmental processes, and to promote targeted therapy for nervous system-related diseases.
Collapse
Affiliation(s)
- Lingling Jiang
- Hengyang Medical College, University of South China, Hengyang, China
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiaoheng Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shasha Wang
- Hengyang Medical College, University of South China, Hengyang, China
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Jinbo Cheng, ; Zengqiang Yuan, ,
| | - Jinbo Cheng
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
- *Correspondence: Jinbo Cheng, ; Zengqiang Yuan, ,
| |
Collapse
|
6
|
Personalized Management and Treatment of Alzheimer's Disease. Life (Basel) 2022; 12:life12030460. [PMID: 35330211 PMCID: PMC8951963 DOI: 10.3390/life12030460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a priority health problem with a high cost to society and a large consumption of medical and social resources. The management of AD patients is complex and multidisciplinary. Over 90% of patients suffer from concomitant diseases and require personalized therapeutic regimens to reduce adverse drug reactions (ADRs), drug−drug interactions (DDIs), and unnecessary costs. Men and women show substantial differences in their AD-related phenotypes. Genomic, epigenetic, neuroimaging, and biochemical biomarkers are useful for predictive and differential diagnosis. The most frequent concomitant diseases include hypertension (>25%), obesity (>70%), diabetes mellitus type 2 (>25%), hypercholesterolemia (40%), hypertriglyceridemia (20%), metabolic syndrome (20%), hepatobiliary disorder (15%), endocrine/metabolic disorders (>20%), cardiovascular disorder (40%), cerebrovascular disorder (60−90%), neuropsychiatric disorders (60−90%), and cancer (10%). Over 90% of AD patients require multifactorial treatments with risk of ADRs and DDIs. The implementation of pharmacogenetics in clinical practice can help optimize the limited therapeutic resources available to treat AD and personalize the use of anti-dementia drugs, in combination with other medications, for the treatment of concomitant disorders.
Collapse
|
7
|
Meng D, Mohammadi-Nejad AR, Sotiropoulos SN, Auer DP. Anticholinergic drugs and forebrain magnetic resonance imaging changes in cognitively normal people and those with mild cognitive impairment. Eur J Neurol 2022; 29:1344-1353. [PMID: 35129272 PMCID: PMC9304308 DOI: 10.1111/ene.15251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/13/2022] [Indexed: 01/24/2023]
Abstract
Background and purpose Anticholinergic (AC) medication use is associated with cognitive decline and dementia, which may be related to an AC‐induced central hypocholinergic state, but the exact mechanisms remain to be understood. We aimed to further elucidate the putative link between AC drug prescription, cognition, and structural and functional impairment of the forebrain cholinergic nucleus basalis of Meynert (NBM). Methods Cognitively normal (CN; n = 344) and mildly cognitively impaired (MCI; n = 224) Alzheimer’s Disease Neuroimaging Initiative Phase 3 participants with good quality 3‐T magnetic resonance imaging were included. Structural (regional gray matter [GM] density) and functional NBM integrity (functional connectivity [FC]) were compared between those on AC medication for > 1 year (AC+) and those without (AC−) in each condition. AC burden was classed as mild, moderate, or severe. Results MCI AC+ participants (0.55 ± 0.03) showed lower NBM GM density compared to MCI AC− participants (0.56 ± 0.03, p = 0.002), but there was no structural AC effect in CN. NBM FC was lower in CN AC+ versus CN AC− (3.6 ± 0.5 vs. 3.9 ± 0.6, p = 0.001), and in MCI AC+ versus MCI AC− (3.3 ± 0.2 vs. 3.7 ± 0.5, p < 0.001), with larger effect size in MCI. NBM FC partially mediated the association between AC medication burden and cognition. Conclusions Our findings provide novel support for a detrimental effect of mild AC medication on the forebrain cholinergic system characterized as functional central hypocholinergic that partially mediated AC‐related cognitive impairment. Moreover, structural tissue damage suggests neurodegeneration, and larger effect sizes in MCI point to enhanced susceptibility for AC medication in those at risk of dementia.
Collapse
Affiliation(s)
- Dewen Meng
- Radiological Sciences, Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.,National Institute for Health Research Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Ali-Reza Mohammadi-Nejad
- Radiological Sciences, Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.,National Institute for Health Research Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Stamatios N Sotiropoulos
- Radiological Sciences, Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.,National Institute for Health Research Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Dorothee P Auer
- Radiological Sciences, Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.,National Institute for Health Research Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
8
|
Cacabelos R, Naidoo V, Martínez-Iglesias O, Corzo L, Cacabelos N, Pego R, Carril JC. Pharmacogenomics of Alzheimer's Disease: Novel Strategies for Drug Utilization and Development. Methods Mol Biol 2022; 2547:275-387. [PMID: 36068470 DOI: 10.1007/978-1-0716-2573-6_13] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD) is a priority health problem in developed countries with a high cost to society. Approximately 20% of direct costs are associated with pharmacological treatment. Over 90% of patients require multifactorial treatments, with risk of adverse drug reactions (ADRs) and drug-drug interactions (DDIs) for the treatment of concomitant diseases such as hypertension (>25%), obesity (>70%), diabetes mellitus type 2 (>25%), hypercholesterolemia (40%), hypertriglyceridemia (20%), metabolic syndrome (20%), hepatobiliary disorder (15%), endocrine/metabolic disorders (>20%), cardiovascular disorder (40%), cerebrovascular disorder (60-90%), neuropsychiatric disorders (60-90%), and cancer (10%).For the past decades, pharmacological studies in search of potential treatments for AD focused on the following categories: neurotransmitter enhancers (11.38%), multitarget drugs (2.45%), anti-amyloid agents (13.30%), anti-tau agents (2.03%), natural products and derivatives (25.58%), novel synthetic drugs (8.13%), novel targets (5.66%), repository drugs (11.77%), anti-inflammatory drugs (1.20%), neuroprotective peptides (1.25%), stem cell therapy (1.85%), nanocarriers/nanotherapeutics (1.52%), and other compounds (<1%).Pharmacogenetic studies have shown that the therapeutic response to drugs in AD is genotype-specific in close association with the gene clusters that constitute the pharmacogenetic machinery (pathogenic, mechanistic, metabolic, transporter, pleiotropic genes) under the regulatory control of epigenetic mechanisms (DNA methylation, histone/chromatin remodeling, microRNA regulation). Most AD patients (>60%) are carriers of over ten pathogenic genes. The genes that most frequently (>50%) accumulate pathogenic variants in the same AD case are A2M (54.38%), ACE (78.94%), BIN1 (57.89%), CLU (63.15%), CPZ (63.15%), LHFPL6 (52.63%), MS4A4E (50.87%), MS4A6A (63.15%), PICALM (54.38%), PRNP (80.7059), and PSEN1 (77.19%). There is also an accumulation of 15 to 26 defective pharmagenes in approximately 85% of AD patients. About 50% of AD patients are carriers of at least 20 mutant pharmagenes, and over 80% are deficient metabolizers for the most common drugs, which are metabolized via the CYP2D6, CYP2C9, CYP2C19, and CYP3A4/5 enzymes.The implementation of pharmacogenetics can help optimize drug development and the limited therapeutic resources available to treat AD, and personalize the use of anti-dementia drugs in combination with other medications for the treatment of concomitant disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain.
| | - Vinogran Naidoo
- Department of Neuroscience, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Olaia Martínez-Iglesias
- Department of Medical Epigenetics, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Lola Corzo
- Department of Medical Biochemistry, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Natalia Cacabelos
- Department of Medical Documentation, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Rocío Pego
- Department of Neuropsychology, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Juan C Carril
- Department of Genomics and Pharmacogenomics, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| |
Collapse
|
9
|
Thibault PA, Ganesan A, Kalyaanamoorthy S, Clarke JPWE, Salapa HE, Levin MC. hnRNP A/B Proteins: An Encyclopedic Assessment of Their Roles in Homeostasis and Disease. BIOLOGY 2021; 10:biology10080712. [PMID: 34439945 PMCID: PMC8389229 DOI: 10.3390/biology10080712] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022]
Abstract
The hnRNP A/B family of proteins is canonically central to cellular RNA metabolism, but due to their highly conserved nature, the functional differences between hnRNP A1, A2/B1, A0, and A3 are often overlooked. In this review, we explore and identify the shared and disparate homeostatic and disease-related functions of the hnRNP A/B family proteins, highlighting areas where the proteins have not been clearly differentiated. Herein, we provide a comprehensive assembly of the literature on these proteins. We find that there are critical gaps in our grasp of A/B proteins' alternative splice isoforms, structures, regulation, and tissue and cell-type-specific functions, and propose that future mechanistic research integrating multiple A/B proteins will significantly improve our understanding of how this essential protein family contributes to cell homeostasis and disease.
Collapse
Affiliation(s)
- Patricia A. Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; (P.A.T.); (J.-P.W.E.C.); (H.E.S.)
- Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Aravindhan Ganesan
- ArGan’s Lab, School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Subha Kalyaanamoorthy
- Department of Chemistry, Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Joseph-Patrick W. E. Clarke
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; (P.A.T.); (J.-P.W.E.C.); (H.E.S.)
- Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Hannah E. Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; (P.A.T.); (J.-P.W.E.C.); (H.E.S.)
- Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Michael C. Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; (P.A.T.); (J.-P.W.E.C.); (H.E.S.)
- Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
- Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Correspondence:
| |
Collapse
|
10
|
Binning W, Hogan-Cann AE, Yae Sakae D, Maksoud M, Ostapchenko V, Al-Onaizi M, Matovic S, Lu WY, Prado MAM, Inoue W, Prado VF. Chronic hM3Dq signaling in microglia ameliorates neuroinflammation in male mice. Brain Behav Immun 2020; 88:791-801. [PMID: 32434046 DOI: 10.1016/j.bbi.2020.05.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
Microglia express muscarinic G protein-coupled receptors (GPCRs) that sense cholinergic activity and are activated by acetylcholine to potentially regulate microglial functions. Knowledge about how distinct types of muscarinic GPCR signaling regulate microglia function in vivo is still poor, partly due to the fact that some of these receptors are also present in astrocytes and neurons. We generated mice expressing the hM3Dq Designer Receptor Exclusively Activated by Designer Drugs (DREADD) selectively in microglia to investigate the role of muscarinic M3Gq-linked signaling. We show that activation of hM3Dq using clozapine N-oxide (CNO) elevated intracellular calcium levels and increased phagocytosis of FluoSpheres by microglia in vitro. Interestingly, whereas acute treatment with CNO increased synthesis of cytokine mRNA, chronic treatment attenuated LPS-induced cytokine mRNA changes in the brain. No effect of CNO on cytokine expression was observed in DREADD-negative mice. Interestingly, CNO activation of M3Dq in microglia was able to attenuate LPS-mediated decrease in social interactions. These results suggest that chronic activation of M3 muscarinic receptors (the hM3Dq progenitor) in microglia, and potentially other Gq-coupled GPCRs, can trigger an inflammatory-like response that preconditions microglia to decrease their response to further immunological challenges. Our results indicate that hM3Dq can be a useful tool to modulate neuroinflammation and study microglial immunological memory in vivo, which may be applicable for manipulations of neuroinflammation in neurodegenerative and psychiatric diseases.
Collapse
Affiliation(s)
- William Binning
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Aja E Hogan-Cann
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Diana Yae Sakae
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Matthew Maksoud
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Valeriy Ostapchenko
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Mohammed Al-Onaizi
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Sara Matovic
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Wei-Yang Lu
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Marco A M Prado
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Anatomy & Cell Biology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada.
| | - Wataru Inoue
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada.
| | - Vania F Prado
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Anatomy & Cell Biology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada.
| |
Collapse
|
11
|
Liu Y, Shi SL. The roles of hnRNP A2/B1 in RNA biology and disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2020; 12:e1612. [PMID: 32588964 DOI: 10.1002/wrna.1612] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022]
Abstract
The RNA-binding protein hnRNPA2/B1 is a member of the hnRNPs family and is widely expressed in various tissues. hnRNPA2/B1 recognizes and binds specific RNA substrates and DNA motifs and is involved in the transcription, splicing processing, transport, stability, and translation regulation of a variety of RNA molecules and in regulating the expression of a large number of genes. hnRNPA2/B1 is also involved in telomere maintenance and DNA repair, while its expression changes and mutations are involved in the development of various tumors and neurodegenerative and autoimmune diseases. This paper reviews the role and mechanism of hnRNPA2/B1 in RNA metabolism, tumors, and neurodegenerative and autoimmune diseases. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Yu Liu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.,School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Song-Lin Shi
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
12
|
Cacabelos R. Pharmacogenetic considerations when prescribing cholinesterase inhibitors for the treatment of Alzheimer's disease. Expert Opin Drug Metab Toxicol 2020; 16:673-701. [PMID: 32520597 DOI: 10.1080/17425255.2020.1779700] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Cholinergic dysfunction, demonstrated in the late 1970s and early 1980s, led to the introduction of acetylcholinesterase inhibitors (AChEIs) in 1993 (Tacrine) to enhance cholinergic neurotransmission as the first line of treatment against Alzheimer's disease (AD). The new generation of AChEIs, represented by Donepezil (1996), Galantamine (2001) and Rivastigmine (2002), is the only treatment for AD to date, together with Memantine (2003). AChEIs are not devoid of side-effects and their cost-effectiveness is limited. An option to optimize the correct use of AChEIs is the implementation of pharmacogenetics (PGx) in the clinical practice. AREAS COVERED (i) The cholinergic system in AD, (ii) principles of AD PGx, (iii) PGx of Donepezil, Galantamine, Rivastigmine, Huperzine and other treatments, and (iv) practical recommendations. EXPERT OPINION The most relevant genes influencing AChEI efficacy and safety are APOE and CYPs. APOE-4 carriers are the worst responders to AChEIs. With the exception of Rivastigmine (UGT2B7, BCHE-K), the other AChEIs are primarily metabolized via CYP2D6, CYP3A4, and UGT enzymes, with involvement of ABC transporters and cholinergic genes (CHAT, ACHE, BCHE, SLC5A7, SLC18A3, CHRNA7) in most ethnic groups. Defective variants may affect the clinical response to AChEIs. PGx geno-phenotyping is highly recommended prior to treatment.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine , Bergondo, Corunna, Spain
| |
Collapse
|
13
|
Wang J, Yang C, Wang H, Li D, Li T, Sun Y, Zhao M, Ma J, Hua W, Yang Z. A New Rat Model of Chronic Cerebral Hypoperfusion Resulting in Early-Stage Vascular Cognitive Impairment. Front Aging Neurosci 2020; 12:86. [PMID: 32351379 PMCID: PMC7174718 DOI: 10.3389/fnagi.2020.00086] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Objective Currently, most models of vascular cognitive impairment are established by occluding the carotid arteries uni- or bilaterally to reduce the cerebral blood flow mimicking chronic cerebral hypoxia. Due to the sudden blood flow interruption, a gradual narrowing of the carotid artery cannot be completely imitated. This paper aims to establish a bilateral carotid stenosis model with mild cognitive dysfunction and mild white matter changes to simulate patients with vascular predementia. Methods Aged Wistar rats (18 months old) underwent either bilateral common carotid artery stenosis (BCAS) or occlusion (BCAO) surgery or a sham operation (control group). The cerebral blood flow in the frontal cortex was measured using Doppler flowmetry. Thirty days after surgery, cognitive function impairments were determined with the Morris water maze; cerebral magnetic resonance imaging was performed to detect changes in fractional anisotropy to assess white matter injuries, and histological studies were performed. Results The aged rats in the BCAS group showed a more gradual cerebral blood flow reduction and a lower mortality rate (11%) compared to rats in the BCAO group. The water maze test revealed a more marginal impairment affecting spatial learning and memory in rats with BCAS than in rats with BCAO. Diffusion tensor imaging detected white matter injuries in the hippocampus and cerebral cortex of BCAS rats. Particularly, a small portion of nerve fibers of the lateral somatosensory cortex was significantly different between rats of the BCAO and BCAS groups. In the BCAS group, the microscopic structure of the hippocampal CA1 region changed slightly after 30 days and sustained a slight mitochondrial crista crack. Fluorescence staining indicated that the number of GFAP-positive cells was increased in rat brains of the BCAS group, and this phenomenon was even more pronounced in the BCAO group. The hnRNPA2/B1 and GABAAR-α1 expression levels were significantly decreased in the hippocampus of rats with BCAS compared to those of controls. Conclusion Severe bilateral carotid stenosis induced mild cognitive dysfunction and slight structural changes in the brains of aged rats. Thus, a chronic cerebral hypoperfusion model was successfully established.
Collapse
Affiliation(s)
- Jinxin Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Chenyi Yang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Haiyun Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China.,Medical College of Nankai University, Nankai University, Tianjin, China.,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Dongxue Li
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Tang Li
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Yi Sun
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Mingshu Zhao
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Ji Ma
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Wei Hua
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Zhuo Yang
- Medical College of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
14
|
Weskamp K, Tank EM, Miguez R, McBride JP, Gómez NB, White M, Lin Z, Gonzalez CM, Serio A, Sreedharan J, Barmada SJ. Shortened TDP43 isoforms upregulated by neuronal hyperactivity drive TDP43 pathology in ALS. J Clin Invest 2020; 130:1139-1155. [PMID: 31714900 PMCID: PMC7269575 DOI: 10.1172/jci130988] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Cortical hyperexcitability and mislocalization of the RNA-binding protein TDP43 are highly conserved features in amyotrophic lateral sclerosis (ALS). Nevertheless, the relationship between these phenomena remains poorly defined. Here, we showed that hyperexcitability recapitulates TDP43 pathology by upregulating shortened TDP43 (sTDP43) splice isoforms. These truncated isoforms accumulated in the cytoplasm and formed insoluble inclusions that sequestered full-length TDP43 via preserved N-terminal interactions. Consistent with these findings, sTDP43 overexpression was toxic to mammalian neurons, suggesting neurodegeneration arising from complementary gain- and loss-of-function mechanisms. In humans and mice, sTDP43 transcripts were enriched in vulnerable motor neurons, and we observed a striking accumulation of sTDP43 within neurons and glia of ALS patients. Collectively, these studies uncover a pathogenic role for alternative TDP43 isoforms in ALS, and implicate sTDP43 as a key contributor to the susceptibility of motor neurons in this disorder.
Collapse
Affiliation(s)
| | | | | | - Jonathon P. McBride
- Department of Neurology
- Cellular and Molecular Biology Program, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicolás B. Gómez
- Department of Neurology
- Cellular and Molecular Biology Program, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Ziqiang Lin
- Department of Basic and Clinical Neuroscience and
| | - Carmen Moreno Gonzalez
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | - Andrea Serio
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | | | - Sami J. Barmada
- Department of Neurology
- Neuroscience Graduate Program, and
- Cellular and Molecular Biology Program, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
15
|
Janickova H, Kljakic O, Rosborough K, Raulic S, Matovic S, Gros R, Saksida LM, Bussey TJ, Inoue W, Prado VF, Prado MAM. Selective decrease of cholinergic signaling from pedunculopontine and laterodorsal tegmental nuclei has little impact on cognition but markedly increases susceptibility to stress. FASEB J 2019; 33:7018-7036. [PMID: 30857416 DOI: 10.1096/fj.201802108r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The pedunculopontine tegmental nucleus (PPT) and laterodorsal tegmental nucleus (LDT) are heterogeneous brainstem structures that contain cholinergic, glutamatergic, and GABAergic neurons. PPT/LDT neurons are suggested to modulate both cognitive and noncognitive functions, yet the extent to which acetylcholine (ACh) signaling from the PPT/LDT is necessary for normal behavior remains uncertain. We addressed this issue by using a mouse model in which PPT/LDT cholinergic signaling is highly decreased by selective deletion of the vesicular ACh transporter (VAChT) gene. This approach interferes exclusively with ACh signaling, leaving signaling by other neurotransmitters from PPT/LDT cholinergic neurons intact and sparing other cells. VAChT mutants were examined on different PPT/LDT-associated cognitive domains. Interestingly, VAChT mutants showed no attentional deficits and only minor cognitive flexibility impairments while presenting large deficiencies in both spatial and cued Morris water maze (MWM) tasks. Conversely, working spatial memory determined with the Y-maze and spatial memory measured with the Barnes maze were not affected, suggesting that deficits in MWM were unrelated to spatial memory abnormalities. Supporting this interpretation, VAChT mutants exhibited alterations in anxiety-like behavior and increased corticosterone levels after exposure to the MWM, suggesting altered stress response. Thus, PPT/LDT VAChT-mutant mice present little cognitive impairment per se, yet they exhibit increased susceptibility to stress, which may lead to performance deficits in more stressful conditions.-Janickova, H., Kljakic, O., Rosborough, K., Raulic, S., Matovic, S., Gros, R., Saksida, L. M., Bussey, T. J., Inoue, W., Prado, V. F., Prado, M. A. M. Selective decrease of cholinergic signaling from pedunculopontine and laterodorsal tegmental nuclei has little impact on cognition but markedly increases susceptibility to stress.
Collapse
Affiliation(s)
- Helena Janickova
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Ornela Kljakic
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Kaie Rosborough
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sanda Raulic
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sara Matovic
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and
| | - Robert Gros
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and
| | - Lisa M Saksida
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada
| | - Timothy J Bussey
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada
| | - Wataru Inoue
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and
| | - Vania F Prado
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; and.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
16
|
Qian L, Milne MR, Shepheard S, Rogers ML, Medeiros R, Coulson EJ. Removal of p75 Neurotrophin Receptor Expression from Cholinergic Basal Forebrain Neurons Reduces Amyloid-β Plaque Deposition and Cognitive Impairment in Aged APP/PS1 Mice. Mol Neurobiol 2018; 56:4639-4652. [DOI: 10.1007/s12035-018-1404-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
|
17
|
Longitudinal Alzheimer’s Degeneration Reflects the Spatial Topography of Cholinergic Basal Forebrain Projections. Cell Rep 2018; 24:38-46. [DOI: 10.1016/j.celrep.2018.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/09/2018] [Accepted: 05/30/2018] [Indexed: 10/28/2022] Open
|
18
|
Turnbull MT, Boskovic Z, Coulson EJ. Acute Down-regulation of BDNF Signaling Does Not Replicate Exacerbated Amyloid-β Levels and Cognitive Impairment Induced by Cholinergic Basal Forebrain Lesion. Front Mol Neurosci 2018. [PMID: 29520217 PMCID: PMC5827359 DOI: 10.3389/fnmol.2018.00051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Degeneration of basal forebrain cholinergic neurons (BFCNs) precedes hippocampal degeneration and pathological amyloid-beta (Aβ) accumulation, and underpins the development of cognitive dysfunction in sporadic Alzheimer’s disease (AD). We hypothesized that degeneration of BFCNs causes a decrease in neurotrophin levels in innervated brain areas, which in turn promotes the development of Aβ pathology and cognitive impairment. Here we show that lesion of septo-hippocampal BFCNs in a pre-symptomatic transgenic amyloid AD mouse model (APP/PS1 mice) increases soluble Aβ levels in the hippocampus, and induces cognitive deficits in a spatial memory task that are not seen in either unlesioned APP/PS1 or non-transgenic littermate control mice. Furthermore, the BFCN lesion results in decreased levels of brain-derived neurotrophic factor (BDNF). However, viral knockdown of neuronal BDNF in the hippocampus of APP/PS1 mice (in the absence of BFCN loss) neither increased the level of Aβ nor caused cognitive deficits. These results suggest that the cognitive decline and Aβ pathology induced by BFCN loss occur independent of dysfunctional neuronal BDNF signaling, and may therefore be directly underpinned by reduced cholinergic neurotransmission.
Collapse
Affiliation(s)
- Marion T Turnbull
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Zoran Boskovic
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Elizabeth J Coulson
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.,Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
19
|
HRPU-2, a Homolog of Mammalian hnRNP U, Regulates Synaptic Transmission by Controlling the Expression of SLO-2 Potassium Channel in Caenorhabditis elegans. J Neurosci 2017; 38:1073-1084. [PMID: 29217678 DOI: 10.1523/jneurosci.1991-17.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/27/2017] [Accepted: 12/02/2017] [Indexed: 12/22/2022] Open
Abstract
Slo2 channels are large-conductance potassium channels abundantly expressed in the nervous system. However, it is unclear how their expression level in neurons is regulated. Here we report that HRPU-2, an RNA-binding protein homologous to mammalian heterogeneous nuclear ribonucleoprotein U (hnRNP U), plays an important role in regulating the expression of SLO-2 (a homolog of mammalian Slo2) in Caenorhabditis elegans Loss-of-function (lf) mutants of hrpu-2 were isolated in a genetic screen for suppressors of a sluggish phenotype caused by a hyperactive SLO-2. In hrpu-2(lf) mutants, SLO-2-mediated delayed outward currents in neurons are greatly decreased, and neuromuscular synaptic transmission is enhanced. These mutant phenotypes can be rescued by expressing wild-type HRPU-2 in neurons. HRPU-2 binds to slo-2 mRNA, and hrpu-2(lf) mutants show decreased SLO-2 protein expression. In contrast, hrpu-2(lf) does not alter the expression of either the BK channel SLO-1 or the Shaker type potassium channel SHK-1. hrpu-2(lf) mutants are indistinguishable from wild type in gross motor neuron morphology and locomotion behavior. Together, these observations suggest that HRPU-2 plays important roles in SLO-2 function by regulating SLO-2 protein expression, and that SLO-2 is likely among a restricted set of proteins regulated by HRPU-2. Mutations of human Slo2 channel and hnRNP U are strongly linked to epileptic disorders and intellectual disability. The findings of this study suggest a potential link between these two molecules in human patients.SIGNIFICANCE STATEMENT Heterogeneous nuclear ribonucleoprotein U (hnRNP U) belongs to a family of RNA-binding proteins that play important roles in controlling gene expression. Recent studies have established a strong link between mutations of hnRNP U and human epilepsies and intellectual disability. However, it is unclear how mutations of hnRNP U may cause such disorders. This study shows that mutations of HRPU-2, a worm homolog of mammalian hnRNP U, result in dysfunction of a Slo2 potassium channel, which is critical to neuronal function. Because mutations of Slo2 channels are also strongly associated with epileptic encephalopathies and intellectual disability in humans, the findings of this study point to a potential mechanism underlying neurological disorders caused by hnRNP U mutations.
Collapse
|
20
|
Kapeli K, Martinez FJ, Yeo GW. Genetic mutations in RNA-binding proteins and their roles in ALS. Hum Genet 2017; 136:1193-1214. [PMID: 28762175 PMCID: PMC5602095 DOI: 10.1007/s00439-017-1830-7] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
Mutations in genes that encode RNA-binding proteins (RBPs) have emerged as critical determinants of neurological diseases, especially motor neuron disorders such as amyotrophic lateral sclerosis (ALS). RBPs are involved in all aspects of RNA processing, controlling the life cycle of RNAs from synthesis to degradation. Hallmark features of RBPs in neuron dysfunction include misregulation of RNA processing, mislocalization of RBPs to the cytoplasm, and abnormal aggregation of RBPs. Much progress has been made in understanding how ALS-associated mutations in RBPs drive pathogenesis. Here, we focus on several key RBPs involved in ALS—TDP-43, HNRNP A2/B1, HNRNP A1, FUS, EWSR1, and TAF15—and review our current understanding of how mutations in these proteins cause disease.
Collapse
Affiliation(s)
- Katannya Kapeli
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Fernando J Martinez
- Department of Cellular and Molecular Medicine, Stem Cell Program and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gene W Yeo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
- Department of Cellular and Molecular Medicine, Stem Cell Program and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
- Molecular Engineering Laboratory, A*STAR, Singapore, 138673, Singapore.
| |
Collapse
|
21
|
Janickova H, Prado VF, Prado MAM, El Mestikawy S, Bernard V. Vesicular acetylcholine transporter (VAChT) over-expression induces major modifications of striatal cholinergic interneuron morphology and function. J Neurochem 2017. [DOI: 10.1111/jnc.14105] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Helena Janickova
- Department of Physiology and Pharmacology and Department of Anatomy & Cell Biology; Robarts Research Institute; Molecular Medicine Laboratories; The University of Western Ontario; London Ontario Canada
| | - Vania F. Prado
- Department of Physiology and Pharmacology and Department of Anatomy & Cell Biology; Robarts Research Institute; Molecular Medicine Laboratories; The University of Western Ontario; London Ontario Canada
| | - Marco A. M. Prado
- Department of Physiology and Pharmacology and Department of Anatomy & Cell Biology; Robarts Research Institute; Molecular Medicine Laboratories; The University of Western Ontario; London Ontario Canada
| | - Salah El Mestikawy
- Sorbonne Universités; Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130; Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS); Paris France
- Department of Psychiatry; Douglas Mental Health University Institute; McGill University; Montreal Canada
| | - Véronique Bernard
- Sorbonne Universités; Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130; Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS); Paris France
| |
Collapse
|
22
|
Kolisnyk B, Al-Onaizi M, Soreq L, Barbash S, Bekenstein U, Haberman N, Hanin G, Kish MT, Souza da Silva J, Fahnestock M, Ule J, Soreq H, Prado VF, Prado MAM. Cholinergic Surveillance over Hippocampal RNA Metabolism and Alzheimer's-Like Pathology. Cereb Cortex 2017; 27:3553-3567. [PMID: 27312991 DOI: 10.1093/cercor/bhw177] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The relationship between long-term cholinergic dysfunction and risk of developing dementia is poorly understood. Here we used mice with deletion of the vesicular acetylcholine transporter (VAChT) in the forebrain to model cholinergic abnormalities observed in dementia. Whole-genome RNA sequencing of hippocampal samples revealed that cholinergic failure causes changes in RNA metabolism. Remarkably, key transcripts related to Alzheimer's disease are affected. BACE1, for instance, shows abnormal splicing caused by decreased expression of the splicing regulator hnRNPA2/B1. Resulting BACE1 overexpression leads to increased APP processing and accumulation of soluble Aβ1-42. This is accompanied by age-related increases in GSK3 activation, tau hyperphosphorylation, caspase-3 activation, decreased synaptic markers, increased neuronal death, and deteriorating cognition. Pharmacological inhibition of GSK3 hyperactivation reversed deficits in synaptic markers and tau hyperphosphorylation induced by cholinergic dysfunction, indicating a key role for GSK3 in some of these pathological changes. Interestingly, in human brains there was a high correlation between decreased levels of VAChT and hnRNPA2/B1 levels with increased tau hyperphosphorylation. These results suggest that changes in RNA processing caused by cholinergic loss can facilitate Alzheimer's-like pathology in mice, providing a mechanism by which decreased cholinergic tone may increase risk of dementia.
Collapse
Affiliation(s)
| | - Mohammed Al-Onaizi
- Robarts Research Institute
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A5K8
| | - Lilach Soreq
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Shahar Barbash
- The Edmond and Lily Safra Center for Brain Science and The Silberman Institute of Life Sciences, The Edmond J Safra Campus, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Uriya Bekenstein
- The Edmond and Lily Safra Center for Brain Science and The Silberman Institute of Life Sciences, The Edmond J Safra Campus, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Nejc Haberman
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Geula Hanin
- The Edmond and Lily Safra Center for Brain Science and The Silberman Institute of Life Sciences, The Edmond J Safra Campus, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Maxine T Kish
- Robarts Research Institute
- Department of Physiology and Pharmacology
| | | | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, CanadaL8S 4K1
| | - Jernej Ule
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Hermona Soreq
- The Edmond and Lily Safra Center for Brain Science and The Silberman Institute of Life Sciences, The Edmond J Safra Campus, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Vania F Prado
- Robarts Research Institute
- Graduate Program in Neuroscience
- Department of Physiology and Pharmacology
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A5K8
| | - Marco A M Prado
- Robarts Research Institute
- Graduate Program in Neuroscience
- Department of Physiology and Pharmacology
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A5K8
| |
Collapse
|
23
|
Lackie RE, Maciejewski A, Ostapchenko VG, Marques-Lopes J, Choy WY, Duennwald ML, Prado VF, Prado MAM. The Hsp70/Hsp90 Chaperone Machinery in Neurodegenerative Diseases. Front Neurosci 2017; 11:254. [PMID: 28559789 PMCID: PMC5433227 DOI: 10.3389/fnins.2017.00254] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
The accumulation of misfolded proteins in the human brain is one of the critical features of many neurodegenerative diseases, including Alzheimer's disease (AD). Assembles of beta-amyloid (Aβ) peptide—either soluble (oligomers) or insoluble (plaques) and of tau protein, which form neurofibrillary tangles, are the major hallmarks of AD. Chaperones and co-chaperones regulate protein folding and client maturation, but they also target misfolded or aggregated proteins for refolding or for degradation, mostly by the proteasome. They form an important line of defense against misfolded proteins and are part of the cellular quality control system. The heat shock protein (Hsp) family, particularly Hsp70 and Hsp90, plays a major part in this process and it is well-known to regulate protein misfolding in a variety of diseases, including tau levels and toxicity in AD. However, the role of Hsp90 in regulating protein misfolding is not yet fully understood. For example, knockdown of Hsp90 and its co-chaperones in a Caenorhabditis elegans model of Aβ misfolding leads to increased toxicity. On the other hand, the use of Hsp90 inhibitors in AD mouse models reduces Aβ toxicity, and normalizes synaptic function. Stress-inducible phosphoprotein 1 (STI1), an intracellular co-chaperone, mediates the transfer of clients from Hsp70 to Hsp90. Importantly, STI1 has been shown to regulate aggregation of amyloid-like proteins in yeast. In addition to its intracellular function, STI1 can be secreted by diverse cell types, including astrocytes and microglia and function as a neurotrophic ligand by triggering signaling via the cellular prion protein (PrPC). Extracellular STI1 can prevent Aβ toxic signaling by (i) interfering with Aβ binding to PrPC and (ii) triggering pro-survival signaling cascades. Interestingly, decreased levels of STI1 in C. elegans can also increase toxicity in an amyloid model. In this review, we will discuss the role of intracellular and extracellular STI1 and the Hsp70/Hsp90 chaperone network in mechanisms underlying protein misfolding in neurodegenerative diseases, with particular focus on AD.
Collapse
Affiliation(s)
- Rachel E Lackie
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Program in Neuroscience, University of Western OntarioLondon, ON, Canada
| | - Andrzej Maciejewski
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Department of Biochemistry, University of Western OntarioLondon, ON, Canada
| | - Valeriy G Ostapchenko
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada
| | - Jose Marques-Lopes
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada
| | - Wing-Yiu Choy
- Department of Biochemistry, University of Western OntarioLondon, ON, Canada
| | - Martin L Duennwald
- Department of Pathology and Laboratory Medicine, University of Western OntarioLondon, ON, Canada
| | - Vania F Prado
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Program in Neuroscience, University of Western OntarioLondon, ON, Canada.,Department of Physiology and Pharmacology, University of Western OntarioLondon, ON, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, ON, Canada
| | - Marco A M Prado
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Program in Neuroscience, University of Western OntarioLondon, ON, Canada.,Department of Physiology and Pharmacology, University of Western OntarioLondon, ON, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, ON, Canada
| |
Collapse
|