1
|
Jin J, Doan J, Fernandez C, Nguyen S, Spencer C, Kleschevnikov AM. Early postnatal GABAB antagonist treatment normalizes inhibitory/excitatory balance in neonatal Ts65Dn mice, a genetic model of down syndrome. Exp Neurol 2025; 386:115171. [PMID: 39889878 DOI: 10.1016/j.expneurol.2025.115171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Brain abnormalities in Down syndrome (DS) most rapidly accumulate during the third trimester, a critical period for the formation of neural circuits in the hippocampus and neocortex. In mice, this stage roughly corresponds to the first 2.5 weeks after birth. We hypothesized that enhanced Girk2 channel signaling during this critical period profoundly contributes to the formation of faulty neural circuits in mouse genetic models of DS, with a key feature being an imbalance of excitatory and inhibitory neurotransmission favoring inhibition. Major predictions of this hypothesis were tested. We observed that hippocampal Girk2 levels are enhanced, GABAB/Girk2 signaling efficiency is increased, and intrinsic neuronal excitability of dentate gyrus (DG) granule cells is reduced in neonatal Ts65Dn mice. Given this, we tested if suppressing the enhanced GABAB/Girk2 signaling in the early postnatal period would affect the inhibitory/excitatory (I/E) balance in Ts65Dn mice. Remarkably, GABAB antagonist treatment from postnatal day 2 (P2) to P17 normalized the exaggerated IPSC/EPSC ratio in DG granule cells in Ts65Dn mice. Our findings show that GABAB/Girk2 signaling is increased in neonatal Ts65Dn mice, and that pharmacological suppression of GABAB receptors during the early postnatal period normalizes the I/E balance. These results suggest that early intervention targeting GABAB/Girk2 signaling could be a promising therapeutic approach to mitigate cognitive impairment in DS.
Collapse
Affiliation(s)
- Joshua Jin
- University of California San Diego, La Jolla, CA, United States
| | - James Doan
- University of California San Diego, La Jolla, CA, United States
| | | | - Samuel Nguyen
- University of California San Diego, La Jolla, CA, United States
| | - Cole Spencer
- University of California San Diego, La Jolla, CA, United States
| | | |
Collapse
|
2
|
Luo H, de Velasco EMF, Gansemer B, Frederick M, Aguado C, Luján R, Thayer SA, Wickman K. Amyloid-β oligomers trigger sex-dependent inhibition of GIRK channel activity in hippocampal neurons in mice. Sci Signal 2024; 17:eado4132. [PMID: 39353038 PMCID: PMC11600338 DOI: 10.1126/scisignal.ado4132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by amyloid plaques and cognitive decline, the latter of which is thought to be driven by soluble oligomeric amyloid-β (oAβ). The dysregulation of G protein-gated inwardly rectifying K+ (GIRK; also known as Kir3) channels has been implicated in rodent models of AD. Here, seeking mechanistic insights, we uncovered a sex-dependent facet of GIRK-dependent signaling in AD-related amyloid pathophysiology. Synthetic oAβ1-42 suppressed GIRK-dependent signaling in hippocampal neurons from male mice, but not from female mice. This effect required cellular prion protein, the receptor mGluR5, and production of arachidonic acid by the phospholipase PLA2. Although oAβ suppressed GIRK channel activity only in male hippocampal neurons, intrahippocampal infusion of oAβ or genetic suppression of GIRK channel activity in hippocampal pyramidal neurons impaired performance on a memory test in both male and female mice. Moreover, genetic enhancement of GIRK channel activity in hippocampal pyramidal neurons blocked oAβ-induced cognitive impairment in both male and female mice. In APP/PS1 AD model mice, GIRK-dependent signaling was diminished in hippocampal CA1 pyramidal neurons from only male mice before cognitive deficit was detected. However, enhancing GIRK channel activity rescued cognitive deficits in older APP/PS1 mice of both sexes. Thus, whereas diminished GIRK channel activity contributes to cognitive deficits in male mice with increased oAβ burden, enhancing its activity may have therapeutic potential for both sexes.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Benjamin Gansemer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - McKinzie Frederick
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carolina Aguado
- Synaptic Structure Laboratory, Departmento de Ciencias Médicas, Instituto de Biomedicina, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete 02006, SPAIN
| | - Rafael Luján
- Synaptic Structure Laboratory, Departmento de Ciencias Médicas, Instituto de Biomedicina, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete 02006, SPAIN
| | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
3
|
Luo H, Frederick M, Marron Fernandez de Velasco E, Oltmanns JO, Wright C, Wickman K. Domain-selective and sex-dependent regulation of learning and memory in mice by GIRK channel activity in CA1 pyramidal neurons of the dorsal hippocampus. Learn Mem 2024; 31:a054022. [PMID: 39375002 PMCID: PMC11472235 DOI: 10.1101/lm.054022.124] [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: 05/10/2024] [Accepted: 09/15/2024] [Indexed: 10/09/2024]
Abstract
G protein-gated inwardly rectifying K+ (GIRK) channels mediate the postsynaptic inhibitory effect of many neurotransmitters in the hippocampus and are implicated in neurological disorders characterized by cognitive deficits. Here, we show that enhancement or suppression of GIRK channel activity in dorsal CA1 pyramidal neurons disrupted novel object recognition in mice, without impacting open field activity or avoidance behavior. Contextual fear learning was also unaffected, but extinction of contextual fear was disrupted by suppression of GIRK channel activity in male mice. Thus, the strength of GIRK channel activity in dorsal CA1 pyramidal neurons regulates select cognitive task performance in mice.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - McKinzie Frederick
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | - Courtney Wright
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
4
|
Borgognone A, Casadellà M, Martínez de Lagrán M, Paredes R, Clotet B, Dierssen M, Elizalde-Torrent A. Lamivudine modulates the expression of neurological impairment-related genes and LINE-1 retrotransposons in brain tissues of a Down syndrome mouse model. Front Aging Neurosci 2024; 16:1386944. [PMID: 39100749 PMCID: PMC11294114 DOI: 10.3389/fnagi.2024.1386944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Elevated activity of retrotransposons is increasingly recognized to be implicated in a wide range of neurodegenerative and neurodevelopmental diseases, including Down syndrome (DS), which is the most common chromosomal condition causing intellectual disability globally. Previous research by our group has revealed that treatment with lamivudine, a reverse transcriptase inhibitor, improved neurobehavioral phenotypes and completely rescued hippocampal-dependent recognition memory in a DS mouse model, Ts65Dn. We hypothesized that retrotransposition rates would increase in the Ts65Dn mouse model, and lamivudine could block retrotransposons. We analyzed the differentially expressed long interspersed element-1 (LINE-1 or L1) mapping on MMU16 and 17, and showed for the first time that retrotransposition could be associated with Ts65Dn's pathology, as misregulation of L1 was found in brain tissues associated with trisomy. In the cerebral cortex, 6 out of 26 upregulated L1s in trisomic treated mice were located in the telomeric region of MMU16 near Ttc3, Kcnj6, and Dscam genes. In the hippocampus, one upregulated L1 element in trisomic treated mice was located near the Fgd4 gene on MMU16. Moreover, two downregulated L1s rescued after treatment with lamivudine were located in the intronic region of Nrxn1 (MMU17) and Snhg14 (MMU7), implicated in a variety of neurodegenerative disorders. To gain further insight into the mechanism of this improvement, we here analyzed the gene expression profile in the hippocampus and cerebral cortex of trisomic mice treated and no-treated with lamivudine compared to their wild-type littermates. We found that treatment with lamivudine rescued the expression of 24% of trisomic genes in the cortex (located on mouse chromosome (MMU) 16 and 17) and 15% in the hippocampus (located in the human chromosome 21 orthologous regions), with important DS candidate genes such as App and Ets2, rescued in both regions.
Collapse
Affiliation(s)
| | | | - María Martínez de Lagrán
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Roger Paredes
- IrsiCaixa, Badalona, Spain
- Department of Infeccious Diseases and Immunity, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
- Department of Pathology, Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, United States
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, Badalona, Spain
- Fundació Lluita contra les Infeccions, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa, Badalona, Spain
- Department of Infeccious Diseases and Immunity, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, Badalona, Spain
- Fundació Lluita contra les Infeccions, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Mara Dierssen
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | | |
Collapse
|
5
|
Llambrich S, Tielemans B, Saliën E, Atzori M, Wouters K, Van Bulck V, Platt M, Vanherp L, Gallego Fernandez N, Grau de la Fuente L, Poptani H, Verlinden L, Himmelreich U, Croitor A, Attanasio C, Callaerts-Vegh Z, Gsell W, Martínez-Abadías N, Vande Velde G. Pleiotropic effects of trisomy and pharmacologic modulation on structural, functional, molecular, and genetic systems in a Down syndrome mouse model. eLife 2024; 12:RP89763. [PMID: 38497812 PMCID: PMC10948151 DOI: 10.7554/elife.89763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Down syndrome (DS) is characterized by skeletal and brain structural malformations, cognitive impairment, altered hippocampal metabolite concentration and gene expression imbalance. These alterations were usually investigated separately, and the potential rescuing effects of green tea extracts enriched in epigallocatechin-3-gallate (GTE-EGCG) provided disparate results due to different experimental conditions. We overcame these limitations by conducting the first longitudinal controlled experiment evaluating genotype and GTE-EGCG prenatal chronic treatment effects before and after treatment discontinuation. Our findings revealed that the Ts65Dn mouse model reflected the pleiotropic nature of DS, exhibiting brachycephalic skull, ventriculomegaly, neurodevelopmental delay, hyperactivity, and impaired memory robustness with altered hippocampal metabolite concentration and gene expression. GTE-EGCG treatment modulated most systems simultaneously but did not rescue DS phenotypes. On the contrary, the treatment exacerbated trisomic phenotypes including body weight, tibia microarchitecture, neurodevelopment, adult cognition, and metabolite concentration, not supporting the therapeutic use of GTE-EGCG as a prenatal chronic treatment. Our results highlight the importance of longitudinal experiments assessing the co-modulation of multiple systems throughout development when characterizing preclinical models in complex disorders and evaluating the pleiotropic effects and general safety of pharmacological treatments.
Collapse
Affiliation(s)
- Sergi Llambrich
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Birger Tielemans
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Ellen Saliën
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Marta Atzori
- Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Kaat Wouters
- Laboratory of Biological Psychology, KU LeuvenLeuvenBelgium
| | | | - Mark Platt
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of LiverpoolLiverpoolUnited Kingdom
| | - Laure Vanherp
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Nuria Gallego Fernandez
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de BarcelonaBarcelonaSpain
| | - Laura Grau de la Fuente
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de BarcelonaBarcelonaSpain
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of LiverpoolLiverpoolUnited Kingdom
| | - Lieve Verlinden
- Clinical and Experimental Endocrinology, KU LeuvenLeuvenBelgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Anca Croitor
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | | | | | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU LeuvenLeuvenBelgium
| | - Neus Martínez-Abadías
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals (BEECA), Facultat de Biologia, Universitat de BarcelonaBarcelonaSpain
| | | |
Collapse
|
6
|
Faralli A, Fucà E, Lazzaro G, Menghini D, Vicari S, Costanzo F. Transcranial Direct Current Stimulation in neurogenetic syndromes: new treatment perspectives for Down syndrome? Front Cell Neurosci 2024; 18:1328963. [PMID: 38456063 PMCID: PMC10917937 DOI: 10.3389/fncel.2024.1328963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/25/2024] [Indexed: 03/09/2024] Open
Abstract
This perspective review aims to explore the potential neurobiological mechanisms involved in the application of transcranial Direct Current Stimulation (tDCS) for Down syndrome (DS), the leading cause of genetically-based intellectual disability. The neural mechanisms underlying tDCS interventions in genetic disorders, typically characterized by cognitive deficits, are grounded in the concept of brain plasticity. We initially present the neurobiological and functional effects elicited by tDCS applications in enhancing neuroplasticity and in regulating the excitatory/inhibitory balance, both associated with cognitive improvement in the general population. The review begins with evidence on tDCS applications in five neurogenetic disorders, including Rett, Prader-Willi, Phelan-McDermid, and Neurofibromatosis 1 syndromes, as well as DS. Available evidence supports tDCS as a potential intervention tool and underscores the importance of advancing neurobiological research into the mechanisms of tDCS action in these conditions. We then discuss the potential of tDCS as a promising non-invasive strategy to mitigate deficits in plasticity and promote fine-tuning of the excitatory/inhibitory balance in DS, exploring implications for cognitive treatment perspectives in this population.
Collapse
Affiliation(s)
- Alessio Faralli
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Elisa Fucà
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Giulia Lazzaro
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Deny Menghini
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
- Life Sciences and Public Health Department, Catholic University of Sacred Heart, Rome, Italy
| | - Floriana Costanzo
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| |
Collapse
|
7
|
Cruciani F, Aparo A, Brusini L, Combi C, Storti SF, Giugno R, Menegaz G, Boscolo Galazzo I. Identifying the joint signature of brain atrophy and gene variant scores in Alzheimer's Disease. J Biomed Inform 2024; 149:104569. [PMID: 38104851 DOI: 10.1016/j.jbi.2023.104569] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The joint modeling of genetic data and brain imaging information allows for determining the pathophysiological pathways of neurodegenerative diseases such as Alzheimer's disease (AD). This task has typically been approached using mass-univariate methods that rely on a complete set of Single Nucleotide Polymorphisms (SNPs) to assess their association with selected image-derived phenotypes (IDPs). However, such methods are prone to multiple comparisons bias and, most importantly, fail to account for potential cross-feature interactions, resulting in insufficient detection of significant associations. Ways to overcome these limitations while reducing the number of traits aim at conveying genetic information at the gene level and capturing the integrated genetic effects of a set of genetic variants, rather than looking at each SNP individually. Their associations with brain IDPs are still largely unexplored in the current literature, though they can uncover new potential genetic determinants for brain modulations in the AD continuum. In this work, we explored an explainable multivariate model to analyze the genetic basis of the grey matter modulations, relying on the AD Neuroimaging Initiative (ADNI) phase 3 dataset. Cortical thicknesses and subcortical volumes derived from T1-weighted Magnetic Resonance were considered to describe the imaging phenotypes. At the same time the genetic counterpart was represented by gene variant scores extracted by the Sequence Kernel Association Test (SKAT) filtering model. Moreover, transcriptomic analysis was carried on to assess the expression of the resulting genes in the main brain structures as a form of validation. Results highlighted meaningful genotype-phenotype interactionsas defined by three latent components showing a significant difference in the projection scores between patients and controls. Among the significant associations, the model highlighted EPHX1 and BCAS1 gene variant scores involved in neurodegenerative and myelination processes, hence relevant for AD. In particular, the first was associated with decreased subcortical volumes and the second with decreasedtemporal lobe thickness. Noteworthy, BCAS1 is particularly expressed in the dentate gyrus. Overall, the proposed approach allowed capturing genotype-phenotype interactions in a restricted study cohort that was confirmed by transcriptomic analysis, offering insights into the underlying mechanisms of neurodegeneration in AD in line with previous findings and suggesting new potential disease biomarkers.
Collapse
Affiliation(s)
- Federica Cruciani
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
| | - Antonino Aparo
- Department of Computer Science, University of Verona, Verona, Italy
| | - Lorenza Brusini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Carlo Combi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Silvia F Storti
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | - Gloria Menegaz
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|
8
|
Chen X, Feng Y, Quinn RJ, Pountney DL, Richardson DR, Mellick GD, Ma L. Potassium Channels in Parkinson's Disease: Potential Roles in Its Pathogenesis and Innovative Molecular Targets for Treatment. Pharmacol Rev 2023; 75:758-788. [PMID: 36918260 DOI: 10.1124/pharmrev.122.000743] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/05/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the midbrain. The loss of neurons results in a subsequent reduction of dopamine in the striatum, which underlies the core motor symptoms of PD. To date, there are no effective treatments to stop, slow, or reverse the pathologic progression of dopaminergic neurodegeneration. This unfortunate predicament is because of the current early stages in understanding the biologic targets and pathways involved in PD pathogenesis. Ion channels have become emerging targets for new therapeutic development for PD due to their essential roles in neuronal function and neuroinflammation. Potassium channels are the most prominent ion channel family and have been shown to be critically important in PD pathology because of their roles in modulating neuronal excitability, neurotransmitter release, synaptic transmission, and neuroinflammation. In this review, members of the subfamilies of voltage-gated K+ channels, inward rectifying K+ channels, and Ca2+-activated K+ channels are described. Evidence of the role of these channels in PD etiology is discussed together with the latest views on related pathologic mechanisms and their potential as biologic targets for developing neuroprotective drugs for PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is the second most common neurodegenerative disorder, featuring progressive degeneration of dopaminergic neurons in the midbrain. It is a multifactorial disease involving multiple risk factors and complex pathobiological mechanisms. Mounting evidence suggests that ion channels play vital roles in the pathogenesis and progression of PD by regulating neuronal excitability and immune cell function. Therefore, they have become "hot" biological targets for PD, as demonstrated by multiple clinical trials of drug candidates targeting ion channels for PD therapy.
Collapse
Affiliation(s)
- Xiaoyi Chen
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Yunjiang Feng
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Ronald J Quinn
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Dean L Pountney
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Des R Richardson
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - George D Mellick
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Linlin Ma
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| |
Collapse
|
9
|
Popova D, Gameiro-Ros I, Youssef MM, Zalamea P, Morris AD, Prytkova I, Jadali A, Kwan KY, Kamarajan C, Salvatore JE, Xuei X, Chorlian DB, Porjesz B, Kuperman S, Dick DM, Goate A, Edenberg HJ, Tischfield JA, Pang ZP, Slesinger PA, Hart RP. Alcohol reverses the effects of KCNJ6 (GIRK2) noncoding variants on excitability of human glutamatergic neurons. Mol Psychiatry 2023; 28:746-758. [PMID: 36207584 PMCID: PMC9542475 DOI: 10.1038/s41380-022-01818-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 01/19/2023]
Abstract
Synonymous and noncoding single nucleotide polymorphisms (SNPs) in the KCNJ6 gene, encoding G protein-gated inwardly rectifying potassium channel subunit 2 (GIRK2), have been linked with increased electroencephalographic frontal theta event-related oscillations (ERO) in subjects diagnosed with alcohol use disorder (AUD). To identify molecular and cellular mechanisms while retaining the appropriate genetic background, we generated induced excitatory glutamatergic neurons (iN) from iPSCs derived from four AUD-diagnosed subjects with KCNJ6 variants ("Affected: AF") and four control subjects without variants ("Unaffected: UN"). Neurons were analyzed for changes in gene expression, morphology, excitability and physiological properties. Single-cell RNA sequencing suggests that KCNJ6 AF variant neurons have altered patterns of synaptic transmission and cell projection morphogenesis. Results confirm that AF neurons express lower levels of GIRK2, have greater neurite area, and elevated excitability. Interestingly, exposure to intoxicating concentrations of ethanol induces GIRK2 expression and reverses functional effects in AF neurons. Ectopic overexpression of GIRK2 alone mimics the effect of ethanol to normalize induced excitability. We conclude that KCNJ6 variants decrease GIRK2 expression and increase excitability and that this effect can be minimized or reduced with ethanol.
Collapse
Affiliation(s)
- Dina Popova
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
| | - Isabel Gameiro-Ros
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark M Youssef
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Petronio Zalamea
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
| | - Ayeshia D Morris
- Joint Program in Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Iya Prytkova
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Azadeh Jadali
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Kelvin Y Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Chella Kamarajan
- Dept. of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jessica E Salvatore
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Xiaoling Xuei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David B Chorlian
- Dept. of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Bernice Porjesz
- Dept. of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Samuel Kuperman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Danielle M Dick
- Rutgers Addiction Research Center, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Alison Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana Univ School of Medicine, Indianapolis, IN, USA
| | - Jay A Tischfield
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
| | - Zhiping P Pang
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
- Child Health Institute, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald P Hart
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA.
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
10
|
Fernández-Blanco Á, Zamora-Moratalla A, Sabariego-Navarro M, Dierssen M. Defective engram allocation contributes to impaired fear memory performance in Down syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523460. [PMID: 36711850 PMCID: PMC9882045 DOI: 10.1101/2023.01.11.523460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Down syndrome (DS) is the most common genetic form of intellectual disability (ID). The cellular and molecular mechanisms contributing to ID in DS are not completely understood. Recent evidence indicates that a given memory is encoded by sparsely distributed neurons, highly activated during learning, the engram cells. Intriguingly, mechanisms that are of paramount importance for engram formation are impaired in DS. Here we explored engram formation in a DS mouse model, the Ts65Dn and we found a reduced number of engram cells in the dentate gyrus (DG), suggesting reduced neuronal allocation to engrams. We also show that trisomic engram cells present reduced number of mature spines than WT engram cells and their excitability is not enhanced during memory recall. In fact, activation of engram cells using a chemogenetic approach does not recover memory deficits in Ts65Dn. Altogether, our findings suggest that perturbations in engram neurons may play a significant role in memory alterations in DS.
Collapse
|
11
|
Bartesaghi R. Brain circuit pathology in Down syndrome: from neurons to neural networks. Rev Neurosci 2022; 34:365-423. [PMID: 36170842 DOI: 10.1515/revneuro-2022-0067] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/28/2022] [Indexed: 11/15/2022]
Abstract
Down syndrome (DS), a genetic pathology caused by triplication of chromosome 21, is characterized by brain hypotrophy and impairment of cognition starting from infancy. While studies in mouse models of DS have elucidated the major neuroanatomical and neurochemical defects of DS, comparatively fewer investigations have focused on the electrophysiology of the DS brain. Electrical activity is at the basis of brain functioning. Therefore, knowledge of the way in which brain circuits operate in DS is fundamental to understand the causes of behavioral impairment and devise targeted interventions. This review summarizes the state of the art regarding the electrical properties of the DS brain, starting from individual neurons and culminating in signal processing in whole neuronal networks. The reported evidence derives from mouse models of DS and from brain tissues and neurons derived from individuals with DS. EEG data recorded in individuals with DS are also provided as a key tool to understand the impact of brain circuit alterations on global brain activity.
Collapse
Affiliation(s)
- Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
12
|
Kleschevnikov AM. Enhanced GIRK2 channel signaling in Down syndrome: A feasible role in the development of abnormal nascent neural circuits. Front Genet 2022; 13:1006068. [PMID: 36171878 PMCID: PMC9510977 DOI: 10.3389/fgene.2022.1006068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
The most distinctive feature of Down syndrome (DS) is moderate to severe cognitive impairment. Genetic, molecular, and neuronal mechanisms of this complex DS phenotype are currently under intensive investigation. It is becoming increasingly clear that the abnormalities arise from a combination of initial changes caused by triplication of genes on human chromosome 21 (HSA21) and later compensatory adaptations affecting multiple brain systems. Consequently, relatively mild initial cognitive deficits become pronounced with age. This pattern of changes suggests that one approach to improving cognitive function in DS is to target the earliest critical changes, the prevention of which can change the ‘trajectory’ of the brain development and reduce the destructive effects of the secondary alterations. Here, we review the experimental data on the role of KCNJ6 in DS-specific brain abnormalities, focusing on a putative role of this gene in the development of abnormal neural circuits in the hippocampus of genetic mouse models of DS. It is suggested that the prevention of these early abnormalities with pharmacological or genetic means can ameliorate cognitive impairment in DS.
Collapse
|
13
|
Dai Y, Jia P, Zhao Z, Gottlieb A. A Method for Bridging Population-Specific Genotypes to Detect Gene Modules Associated with Alzheimer's Disease. Cells 2022; 11:2219. [PMID: 35883662 PMCID: PMC9319087 DOI: 10.3390/cells11142219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Genome-wide association studies have successfully identified variants associated with multiple conditions. However, generalizing discoveries across diverse populations remains challenging due to large variations in genetic composition. Methods that perform gene expression imputation have attempted to address the transferability of gene discoveries across populations, but with limited success. METHODS Here, we introduce a pipeline that combines gene expression imputation with gene module discovery, including a dense gene module search and a gene set variation analysis, to address the transferability issue. Our method feeds association probabilities of imputed gene expression with a selected phenotype into tissue-specific gene-module discovery over protein interaction networks to create higher-level gene modules. RESULTS We demonstrate our method's utility in three case-control studies of Alzheimer's disease (AD) for three different race/ethnic populations (Whites, African descent and Hispanics). We discovered 182 AD-associated genes from gene modules shared between these populations, highlighting new gene modules associated with AD. CONCLUSIONS Our innovative framework has the potential to identify robust discoveries across populations based on gene modules, as demonstrated in AD.
Collapse
Affiliation(s)
- Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Assaf Gottlieb
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
14
|
Luo H, Marron Fernandez de Velasco E, Wickman K. Neuronal G protein-gated K + channels. Am J Physiol Cell Physiol 2022; 323:C439-C460. [PMID: 35704701 PMCID: PMC9362898 DOI: 10.1152/ajpcell.00102.2022] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels exert a critical inhibitory influence on neurons. Neuronal GIRK channels mediate the G protein-dependent, direct/postsynaptic inhibitory effect of many neurotransmitters including γ-aminobutyric acid (GABA), serotonin, dopamine, adenosine, somatostatin, and enkephalin. In addition to their complex regulation by G proteins, neuronal GIRK channel activity is sensitive to PIP2, phosphorylation, regulator of G protein signaling (RGS) proteins, intracellular Na+ and Ca2+, and cholesterol. The application of genetic and viral manipulations in rodent models, together with recent progress in the development of GIRK channel modulators, has increased our understanding of the physiological and behavioral impact of neuronal GIRK channels. Work in rodent models has also revealed that neuronal GIRK channel activity is modified, transiently or persistently, by various stimuli including exposure drugs of abuse, changes in neuronal activity patterns, and aversive experience. A growing body of preclinical and clinical evidence suggests that dysregulation of GIRK channel activity contributes to neurological diseases and disorders. The primary goals of this review are to highlight fundamental principles of neuronal GIRK channel biology, mechanisms of GIRK channel regulation and plasticity, the nascent landscape of GIRK channel pharmacology, and the potential relevance of GIRK channels to the pathophysiology and treatment of neurological diseases and disorders.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
15
|
Sawa M, Overk C, Becker A, Derse D, Albay R, Weldy K, Salehi A, Beach TG, Doran E, Head E, Yu YE, Mobley WC. Impact of increased APP gene dose in Down syndrome and the Dp16 mouse model. Alzheimers Dement 2022; 18:1203-1234. [PMID: 34757693 PMCID: PMC9085977 DOI: 10.1002/alz.12463] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION People with Down syndrome (DS) are predisposed to Alzheimer's disease (AD). The amyloid hypothesis informs studies of AD. In AD-DS, but not sporadic AD, increased APP copy number is necessary, defining the APP gene dose hypothesis. Which amyloid precursor protein (APP) products contribute needs to be determined. METHODS Brain levels of full-length protein (fl-hAPP), C-terminal fragments (hCTFs), and amyloid beta (Aβ) peptides were measured in DS, AD-DS, non-demented controls (ND), and sporadic AD cases. The APP gene-dose hypothesis was evaluated in the Dp16 model. RESULTS DS and AD-DS differed from ND and AD for all APP products. In AD-DS, Aβ42 and Aβ40 levels exceeded AD. APP products were increased in the Dp16 model; increased APP gene dose was necessary for loss of vulnerable neurons, tau pathology, and activation of astrocytes and microglia. DISCUSSION Increases in APP products other than Aβ distinguished AD-DS from AD. Deciphering AD-DS pathogenesis necessitates deciphering which APP products contribute and how.
Collapse
Affiliation(s)
- Mariko Sawa
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Cassia Overk
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ann Becker
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Dominique Derse
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ricardo Albay
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Kim Weldy
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ahmad Salehi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305
| | - Thomas G. Beach
- Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ 85351
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, 92697
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, 92697
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624,Correspondence to: William Mobley M.D., Department of Neurosciences, UCSD School of Medicine, 9500 Gilman Drive, GPL 355, La Jolla, CA 92093-0624;
| |
Collapse
|
16
|
Hu Y, Liu F, Peng W, Song S, Zhang C, Meng X. Overexpression of miR-99a in hippocampus leads to impairment of reversal learning in mice. Behav Brain Res 2022; 416:113542. [PMID: 34425183 DOI: 10.1016/j.bbr.2021.113542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 11/28/2022]
Abstract
As one of the most common human genetic disorders, Down syndrome (DS) is characterized by a mild-to-moderate cognitive disability, which mainly results from genes overexpression on chromosome 21. The expression of miR-99a, a gene harboring on chromosome 21, is increased by 50 folds in DS brain samples. This study aims to investigate the effect of miR-99a overexpression in the hippocampus on mouse behaviors and explore the underlying mechanisms. Lentivirus vectors were delivered into the hippocampus for focal miR-99a overexpression in mice. Then behaviors were observed by an open field, elevated plus maze, rotarod motor test, and Morris water maze. The genes affected by miR-99a were identified by RNA sequencing (RNA-seq) and confirmed by quantitative RT-PCR (qRT-PCR) in samples isolated from the hippocampus injected with lentivirus-GFP-miR-99a or lentivirus-GFP vectors. It was found that the expression of miR-99a with intrahippocampal delivery of lentivirus-GFP-miR-99a resulted in reversal learning impairment in mice although it had no influence on motor function and anxiety. Meanwhile, RNA-seq results showed that 92 genes including mRNAs and microRNAs were significantly regulated by miR-99a, consistent with qRT-PCR consequence. Moreover, dual-luciferase reporter assay showed that miR-99a could directly bind to the 3'-untranslated regions (3'UTR) of target genes (Clic6 and Kcnj13) with an inhibitory effect on their activity. Furthermore, we also found that miR-99a overexpression affected different biological processes by bioinformatic analyses. Our study showed that miR-99a overexpression in the hippocampus leads to cognitive impairment through regulating the expressions of various genes, which reveals a novel function of miR-99a and provides new insights into understanding the pathophysiologic process of DS.
Collapse
Affiliation(s)
- Yue Hu
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feng Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenpeng Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuxin Song
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
17
|
Bartesaghi R, Vicari S, Mobley WC. Prenatal and Postnatal Pharmacotherapy in Down Syndrome: The Search to Prevent or Ameliorate Neurodevelopmental and Neurodegenerative Disorders. Annu Rev Pharmacol Toxicol 2022; 62:211-233. [PMID: 34990205 PMCID: PMC9632639 DOI: 10.1146/annurev-pharmtox-041521-103641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Those with Down syndrome (DS)-trisomy for chromosome 21-are routinely impacted by cognitive dysfunction and behavioral challenges in children and adults and Alzheimer's disease in older adults. No proven treatments specifically address these cognitive or behavioral changes. However, advances in the establishment of rodent models and human cell models promise to support development of such treatments. A research agenda that emphasizes the identification of overexpressed genes that contribute demonstrably to abnormalities in cognition and behavior in model systems constitutes a rational next step. Normalizing expression of such genes may usher in an era of successful treatments applicable across the life span for those with DS.
Collapse
Affiliation(s)
- Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Stefano Vicari
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, 00168 Rome, Italy,Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165-00146 Rome, Italy
| | - William C. Mobley
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
18
|
Kleschevnikov A. GIRK2 Channels in Down Syndrome and Alzheimer's Disease. Curr Alzheimer Res 2022; 19:819-829. [PMID: 36567290 DOI: 10.2174/1567205020666221223122110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/27/2022]
Abstract
Cognitive impairment in Down syndrome (DS) results from the abnormal expression of hundreds of genes. However, the impact of KCNJ6, a gene located in the middle of the 'Down syndrome critical region' of chromosome 21, seems to stand out. KCNJ6 encodes GIRK2 (KIR3.2) subunits of G protein-gated inwardly rectifying potassium channels, which serve as effectors for GABAB, m2, 5HT1A, A1, and many other postsynaptic metabotropic receptors. GIRK2 subunits are heavily expressed in neocortex, cerebellum, and hippocampus. By controlling resting membrane potential and neuronal excitability, GIRK2 channels may thus affect both synaptic plasticity and stability of neural circuits in the brain regions important for learning and memory. Here, we discuss recent experimental data regarding the role of KCNJ6/GIRK2 in neuronal abnormalities and cognitive impairment in models of DS and Alzheimer's disease (AD). The results compellingly show that signaling through GIRK2 channels is abnormally enhanced in mouse genetic models of Down syndrome and that partial suppression of GIRK2 channels with pharmacological or genetic means can restore synaptic plasticity and improve impaired cognitive functions. On the other hand, signaling through GIRK2 channels is downregulated in AD models, such as models of early amyloidopathy. In these models, reduced GIRK2 channel signaling promotes neuronal hyperactivity, causing excitatory-inhibitory imbalance and neuronal death. Accordingly, activation of GABAB/GIRK2 signaling by GIRK channel activators or GABAB receptor agonists may reduce Aβ-induced hyperactivity and subsequent neuronal death, thereby exerting a neuroprotective effect in models of AD.
Collapse
|
19
|
Abstract
GABAB receptors are implicated in numerous central nervous system-based behaviours and mechanisms, including cognitive processing in preclinical animal models. Homeostatic changes in the expression and function of these receptors across brain structures have been found to affect cognitive processing. Numerous preclinical studies have focused on the role of GABAB receptors in learning, memory and cognition per se with some interesting, although sometimes contradictory, findings. The majority of the existing clinical literature focuses on alterations in GABAB receptor function in conditions and disorders whose main symptomatology includes deficits in cognitive processing. The aim of this chapter is to delineate the role of GABAB receptors in cognitive processes in health and disease of animal models and human clinical populations. More specifically, this review aims to present literature on the role of GABAB receptors in animal models with cognitive deficits, especially those of learning and memory. Further, it aims to capture the progress and advances of research studies on the effects of GABAB receptor compounds in neurodevelopmental and neurodegenerative conditions with cognitive dysfunctions. The neurodevelopmental conditions covered include autism spectrum disorders, fragile X syndrome and Down's syndrome and the neurodegenerative conditions discussed are Alzheimer's disease, epilepsy and autoimmune anti-GABAB encephalitis. Although some findings are contradictory, results indicate a possible therapeutic role of GABAB receptor compounds for the treatment of cognitive dysfunction and learning/memory impairments for some of these conditions, especially in neurodegeneration. Moreover, future research efforts should aim to develop selective GABAB receptor compounds with minimal, if any, side effects.
Collapse
|
20
|
Parrini M, Naskar S, Alberti M, Colombi I, Morelli G, Rocchi A, Nanni M, Piccardi F, Charles S, Ronzitti G, Mingozzi F, Contestabile A, Cancedda L. Restoring neuronal chloride homeostasis with anti-NKCC1 gene therapy rescues cognitive deficits in a mouse model of Down syndrome. Mol Ther 2021; 29:3072-3092. [PMID: 34058387 PMCID: PMC8531145 DOI: 10.1016/j.ymthe.2021.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 01/24/2023] Open
Abstract
A common feature of diverse brain disorders is the alteration of GABA-mediated inhibition because of aberrant, intracellular chloride homeostasis induced by changes in the expression and/or function of chloride transporters. Notably, pharmacological inhibition of the chloride importer NKCC1 is able to rescue brain-related core deficits in animal models of these pathologies and in some human clinical studies. Here, we show that reducing NKCC1 expression by RNA interference in the Ts65Dn mouse model of Down syndrome (DS) restores intracellular chloride concentration, efficacy of gamma-aminobutyric acid (GABA)-mediated inhibition, and neuronal network dynamics in vitro and ex vivo. Importantly, adeno-associated virus (AAV)-mediated, neuron-specific NKCC1 knockdown in vivo rescues cognitive deficits in diverse behavioral tasks in Ts65Dn animals. Our results highlight a mechanistic link between NKCC1 expression and behavioral abnormalities in DS mice and establish a molecular target for new therapeutic approaches, including gene therapy, to treat brain disorders characterized by neuronal chloride imbalance.
Collapse
Affiliation(s)
- Martina Parrini
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Shovan Naskar
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Micol Alberti
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Ilaria Colombi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Giovanni Morelli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Anna Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genoa, Italy; IRCSS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marina Nanni
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Federica Piccardi
- Animal Facility, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Severine Charles
- Genethon, 91000 Evry, France; Paris-Saclay University, University Evry, Inserm, Integrare research unit UMR_S951, 91000 Evry, France
| | - Giuseppe Ronzitti
- Genethon, 91000 Evry, France; Paris-Saclay University, University Evry, Inserm, Integrare research unit UMR_S951, 91000 Evry, France
| | - Federico Mingozzi
- Genethon, 91000 Evry, France; Paris-Saclay University, University Evry, Inserm, Integrare research unit UMR_S951, 91000 Evry, France
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy; Dulbecco Telethon Institute, 00185 Rome, Italy.
| |
Collapse
|
21
|
Chen X, Salehi A, Pearn ML, Overk C, Nguyen PD, Kleschevnikov AM, Maccecchini M, Mobley WC. Targeting increased levels of APP in Down syndrome: Posiphen-mediated reductions in APP and its products reverse endosomal phenotypes in the Ts65Dn mouse model. Alzheimers Dement 2021; 17:271-292. [PMID: 32975365 PMCID: PMC7984396 DOI: 10.1002/alz.12185] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Recent clinical trials targeting amyloid beta (Aβ) and tau in Alzheimer's disease (AD) have yet to demonstrate efficacy. Reviewing the hypotheses for AD pathogenesis and defining possible links between them may enhance insights into both upstream initiating events and downstream mechanisms, thereby promoting discovery of novel treatments. Evidence that in Down syndrome (DS), a population markedly predisposed to develop early onset AD, increased APP gene dose is necessary for both AD neuropathology and dementia points to normalization of the levels of the amyloid precursor protein (APP) and its products as a route to further define AD pathogenesis and discovering novel treatments. BACKGROUND AD and DS share several characteristic manifestations. DS is caused by trisomy of whole or part of chromosome 21; this chromosome contains about 233 protein-coding genes, including APP. Recent evidence points to a defining role for increased expression of the gene for APP and for its 99 amino acid C-terminal fragment (C99, also known as β-CTF) in dysregulating the endosomal/lysosomal system. The latter is critical for normal cellular function and in neurons for transmitting neurotrophic signals. NEW/UPDATED HYPOTHESIS We hypothesize that the increase in APP gene dose in DS initiates a process in which increased levels of full-length APP (fl-APP) and its products, including β-CTF and possibly Aβ peptides (Aβ42 and Aβ40), drive AD pathogenesis through an endosome-dependent mechanism(s), which compromises transport of neurotrophic signals. To test this hypothesis, we carried out studies in the Ts65Dn mouse model of DS and examined the effects of Posiphen, an orally available small molecule shown in prior studies to reduce fl-APP. In vitro, Posiphen lowered fl-APP and its C-terminal fragments, reversed Rab5 hyperactivation and early endosome enlargement, and restored retrograde transport of neurotrophin signaling. In vivo, Posiphen treatment (50 mg/kg/d, 26 days, intraperitoneal [i.p.]) of Ts65Dn mice was well tolerated and demonstrated no adverse effects in behavior. Treatment resulted in normalization of the levels of fl-APP, C-terminal fragments and small reductions in Aβ species, restoration to normal levels of Rab5 activity, reduced phosphorylated tau (p-tau), and reversed deficits in TrkB (tropomyosin receptor kinase B) activation and in the Akt (protein kinase B [PKB]), ERK (extracellular signal-regulated kinase), and CREB (cAMP response element-binding protein) signaling pathways. Remarkably, Posiphen treatment also restored the level of choline acetyltransferase protein to 2N levels. These findings support the APP gene dose hypothesis, point to the need for additional studies to explore the mechanisms by which increased APP gene expression acts to increase the risk for AD in DS, and to possible utility of treatments to normalize the levels of APP and its products for preventing AD in those with DS. MAJOR CHALLENGES FOR THE HYPOTHESIS Important unanswered questions are: (1) When should one intervene in those with DS; (2) would an APP-based strategy have untoward consequences on possible adaptive changes induced by chronically increased APP gene dose; (3) do other genes present on chromosome 21, or on other chromosomes whose expression is dysregulated in DS, contribute to AD pathogenesis; and (4) can one model strategies that combine the use of an APP-based treatment with those directed at other AD phenotypes including p-tau and inflammation. LINKAGE TO OTHER MAJOR THEORIES The APP gene dose hypothesis interfaces with the amyloid cascade hypothesis of AD as well as with the genetic and cell biological observations that support it. Moreover, upregulation of fl-APP protein and products may drive downstream events that dysregulate tau homeostasis and inflammatory responses that contribute to propagation of AD pathogenesis.
Collapse
Affiliation(s)
- Xu‐Qiao Chen
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Ahmad Salehi
- Department of Psychiatry & Behavioral SciencesStanford Medical SchoolPalo AltoCaliforniaUSA
| | - Matthew L. Pearn
- Department of AnesthesiologyUniversity of California San Diego, School of MedicineLa JollaCaliforniaUSA
- V.A. San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - Cassia Overk
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Phuong D. Nguyen
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | | | | | - William C. Mobley
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
22
|
Puente-Bedia A, Berciano MT, Tapia O, Martínez-Cué C, Lafarga M, Rueda N. Nuclear Reorganization in Hippocampal Granule Cell Neurons from a Mouse Model of Down Syndrome: Changes in Chromatin Configuration, Nucleoli and Cajal Bodies. Int J Mol Sci 2021; 22:ijms22031259. [PMID: 33514010 PMCID: PMC7865916 DOI: 10.3390/ijms22031259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 01/05/2023] Open
Abstract
Down syndrome (DS) or trisomy of chromosome 21 (Hsa21) is characterized by impaired hippocampal-dependent learning and memory. These alterations are due to defective neurogenesis and to neuromorphological and functional anomalies of numerous neuronal populations, including hippocampal granular cells (GCs). It has been proposed that the additional gene dose in trisomic cells induces modifications in nuclear compartments and on the chromatin landscape, which could contribute to some DS phenotypes. The Ts65Dn (TS) mouse model of DS carries a triplication of 92 genes orthologous to those found in Hsa21, and shares many phenotypes with DS individuals, including cognitive and neuromorphological alterations. Considering its essential role in hippocampal memory formation, we investigated whether the triplication of this set of Hsa21 orthologous genes in TS mice modifies the nuclear architecture of their GCs. Our results show that the TS mouse presents alterations in the nuclear architecture of its GCs, affecting nuclear compartments involved in transcription and pre-rRNA and pre-mRNA processing. In particular, the GCs of the TS mouse show alterations in the nucleolar fusion pattern and the molecular assembly of Cajal bodies (CBs). Furthermore, hippocampal GCs of TS mice present an epigenetic dysregulation of chromatin that results in an increased heterochromatinization and reduced global transcriptional activity. These nuclear alterations could play an important role in the neuromorphological and/or functional alterations of the hippocampal GCs implicated in the cognitive dysfunction characteristic of TS mice.
Collapse
Affiliation(s)
- Alba Puente-Bedia
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain; (A.P.-B.); (C.M.-C.)
| | - María T. Berciano
- Department of Molecular Biology, “Red sobre Enfermedades Neurodegenerativas (CIBERNED)” and University of Cantabria-IDIVAL, 39011 Santander, Spain;
| | - Olga Tapia
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), “Red sobre Enfermedades Neurodegenerativas (CIBERNED)” and Universidad Europea del Atlántico, 39011 Santander, Spain;
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain; (A.P.-B.); (C.M.-C.)
| | - Miguel Lafarga
- Department of Anatomy and Cell Biology, “Red sobre Enfermedades Neurodegenerativas (CIBERNED)” and University of Cantabria-IDIVAL, 39011 Santander, Spain
- Correspondence: (M.L.); (N.R.); Tel.: +34-942201966 (N.R.); Fax: +34-942201903 (N.R.)
| | - Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain; (A.P.-B.); (C.M.-C.)
- Correspondence: (M.L.); (N.R.); Tel.: +34-942201966 (N.R.); Fax: +34-942201903 (N.R.)
| |
Collapse
|
23
|
Therapeutic potential of targeting G protein-gated inwardly rectifying potassium (GIRK) channels in the central nervous system. Pharmacol Ther 2021; 223:107808. [PMID: 33476640 DOI: 10.1016/j.pharmthera.2021.107808] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
G protein-gated inwardly rectifying potassium channels (Kir3/GirK) are important for maintaining resting membrane potential, cell excitability and inhibitory neurotransmission. Coupled to numerous G protein-coupled receptors (GPCRs), they mediate the effects of many neurotransmitters, neuromodulators and hormones contributing to the general homeostasis and particular synaptic plasticity processes, learning, memory and pain signaling. A growing number of behavioral and genetic studies suggest a critical role for the appropriate functioning of the central nervous system, as well as their involvement in many neurologic and psychiatric conditions, such as neurodegenerative diseases, mood disorders, attention deficit hyperactivity disorder, schizophrenia, epilepsy, alcoholism and drug addiction. Hence, GirK channels emerge as a very promising tool to be targeted in the current scenario where these conditions already are or will become a global public health problem. This review examines recent findings on the physiology, function, dysfunction, and pharmacology of GirK channels in the central nervous system and highlights the relevance of GirK channels as a worthful potential target to improve therapies for related diseases.
Collapse
|
24
|
Shekari A, Fahnestock M. Cholinergic neurodegeneration in Alzheimer disease mouse models. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:191-209. [PMID: 34266592 DOI: 10.1016/b978-0-12-819973-2.00013-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cholinergic signaling is critical for cognitive function. The basal forebrain is the major cholinergic output of the central nervous system. Degeneration of basal forebrain cholinergic neurons is a hallmark of Alzheimer's disease (AD). Mouse models are invaluable tools in disease research and have been used to study AD for over 25 years. However, animal models of AD vary greatly with respect to the degree of cholinergic degeneration observed. The following review will outline the most influential animal models of AD with an emphasis on the basal forebrain cholinergic system.
Collapse
Affiliation(s)
- Arman Shekari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
25
|
Yang W, Sung K, Xu W, Rodriguez MJ, Wu AC, Santos SA, Fang S, Uber RK, Dong SX, Guillory BC, Orain X, Raus J, Jolivalt C, Calcutt N, Rissman RA, Ding J, Wu C. A missense point mutation in nerve growth factor (NGF R100W) results in selective peripheral sensory neuropathy. Prog Neurobiol 2020; 194:101886. [PMID: 32693191 DOI: 10.1016/j.pneurobio.2020.101886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/09/2020] [Accepted: 07/11/2020] [Indexed: 01/15/2023]
Abstract
The R100W mutation in nerve growth factor is associated with hereditary sensory autonomic neuropathy V in a Swedish family. These patients develop severe loss of perception to deep pain but with apparently normal cognitive functions. To better understand the disease mechanism, we examined a knockin mouse model of HSAN V. The homozygous mice showed significant structural deficits in intra-epidermal nerve fibers (IENFs) at birth. These mice had a total loss of pain perception at ∼2 months of age and often failed to survive to adulthood. Heterozygous mutant mice developed a progressive degeneration of small sensory fibers both behaviorally and functionally: they showed a progressive loss of IENFs starting at the age of 9 months accompanied with progressive loss of perception to painful stimuli such as noxious temperature. Quantitative analysis of lumbar 4/5 dorsal root ganglia revealed a significant reduction in small size neurons, while analysis of sciatic nerve fibers revealed the heterozygous mutant mice had no reduction in myelinated nerve fibers. Significantly, the amount of NGF secreted from mouse embryonic fibroblasts were reduced from both heterozygous and homozygous mice compared to their wild-type littermates. Interestingly, the heterozygous mice showed no apparent structural alteration in the brain: neither the anterior cingulate cortex nor the medial septum including NGF-dependent basal forebrain cholinergic neurons. Accordingly, these animals did not develop appreciable deficits in tests for brain function. Our study has thus demonstrated that the NGFR100W mutation likely affects the structure and function of peripheral sensory neurons.
Collapse
Affiliation(s)
- Wanlin Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Neurology, Zhuijiang Hospital, Southern Medical University, Guangzhou, China
| | - Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maria J Rodriguez
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Andrew C Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Sarai A Santos
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Savannah Fang
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Rebecca K Uber
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Stephanie X Dong
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Brandon C Guillory
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Xavier Orain
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jordan Raus
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Corrine Jolivalt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Nigel Calcutt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
26
|
Kessi M, Chen B, Peng J, Tang Y, Olatoutou E, He F, Yang L, Yin F. Intellectual Disability and Potassium Channelopathies: A Systematic Review. Front Genet 2020; 11:614. [PMID: 32655623 PMCID: PMC7324798 DOI: 10.3389/fgene.2020.00614] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/20/2020] [Indexed: 01/15/2023] Open
Abstract
Intellectual disability (ID) manifests prior to adulthood as severe limitations to intellectual function and adaptive behavior. The role of potassium channelopathies in ID is poorly understood. Therefore, we aimed to evaluate the relationship between ID and potassium channelopathies. We hypothesized that potassium channelopathies are strongly associated with ID initiation, and that both gain- and loss-of-function mutations lead to ID. This systematic review explores the burden of potassium channelopathies, possible mechanisms, advancements using animal models, therapies, and existing gaps. The literature search encompassed both PubMed and Embase up to October 2019. A total of 75 articles describing 338 cases were included in this review. Nineteen channelopathies were identified, affecting the following genes: KCNMA1, KCNN3, KCNT1, KCNT2, KCNJ10, KCNJ6, KCNJ11, KCNA2, KCNA4, KCND3, KCNH1, KCNQ2, KCNAB1, KCNQ3, KCNQ5, KCNC1, KCNB1, KCNC3, and KCTD3. Twelve of these genes presented both gain- and loss-of-function properties, three displayed gain-of-function only, three exhibited loss-of-function only, and one had unknown function. How gain- and loss-of-function mutations can both lead to ID remains largely unknown. We identified only a few animal studies that focused on the mechanisms of ID in relation to potassium channelopathies and some of the few available therapeutic options (channel openers or blockers) appear to offer limited efficacy. In conclusion, potassium channelopathies contribute to the initiation of ID in several instances and this review provides a comprehensive overview of which molecular players are involved in some of the most prominent disease phenotypes.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China.,Kilimanjaro Christian Medical University College, Moshi, Tanzania.,Mawenzi Regional Referral Hospital, Moshi, Tanzania
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Yulin Tang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Eleonore Olatoutou
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| |
Collapse
|
27
|
Szczepanik JC, Garcia AF, Lopes de Almeida GR, Cunha MP, Dafre AL. Protective effects against memory impairment induced by methylglyoxal in mice co-treated with FPS-ZM1, an advanced glycation end products receptor antagonist. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
28
|
Szczepanik JC, de Almeida GRL, Cunha MP, Dafre AL. Repeated Methylglyoxal Treatment Depletes Dopamine in the Prefrontal Cortex, and Causes Memory Impairment and Depressive-Like Behavior in Mice. Neurochem Res 2019; 45:354-370. [PMID: 31786717 DOI: 10.1007/s11064-019-02921-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 01/17/2023]
Abstract
Methylglyoxal (MGO) is a highly reactive dicarbonyl molecule that promotes the formation of advanced glycation end products (AGEs), which are believed to play a key role in a number of pathologies, such as diabetes, Alzheimer's disease, and inflammation. Here, Swiss mice were treated with MGO by intraperitoneal injection to investigate its effects on motor activity, mood, and cognition. Acute MGO treatment heavily decreased locomotor activity in the open field test at higher doses (80-200 mg/kg), an effect not observed at lower doses (10-50 mg/kg). Several alterations were observed 4 h after a single MGO injection (10-50 mg/kg): (a) plasma MGO levels were increased, (b) memory was impaired (object location task), (c) anxiolytic behavior was observed in the open field and marble burying test, and (d) depressive-like behavior was evidenced as evaluated by the tail suspension test. Biochemical alterations in the glutathione and glyoxalase systems were not observed 4 h after MGO treatment. Mice were also treated daily with MGO at 0, 10, 25 and 50 mg/kg for 11 days. From the 5th to the 11th day, several behavioral end points were evaluated, resulting in: (a) absence of motor impairment as evaluated in the open field, horizontal bars and pole test, (b) depressive-like behavior observed in the tail suspension test, and (c) cognitive impairments detected on working, short- and long-term memory when mice were tested in the Y-maze spontaneous alternation, object location and recognition tests, and step-down inhibitory avoidance task. An interesting finding was a marked decrease in dopamine levels in the prefrontal cortex of mice treated with 50 mg/kg MGO for 11 days, along with a ~ 25% decrease in the Glo1 content. The MGO-induced dopamine depletion in the prefrontal cortex may be related to the observed memory deficits and depressive-like behavior, an interesting topic to be further studied as a potentially novel route for MGO toxicity.
Collapse
Affiliation(s)
- Jozimar Carlos Szczepanik
- Neurosciences Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Gudrian Ricardo Lopes de Almeida
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Mauricio Peña Cunha
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Alcir Luiz Dafre
- Neurosciences Post-Graduation Program, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
29
|
Unbalanced dendritic inhibition of CA1 neurons drives spatial-memory deficits in the Ts2Cje Down syndrome model. Nat Commun 2019; 10:4991. [PMID: 31676751 PMCID: PMC6825203 DOI: 10.1038/s41467-019-13004-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Overinhibition is assumed one of the main causes of cognitive deficits (e.g. memory impairment) in mouse models of Down syndrome (DS). Yet the mechanisms that drive such exaggerated synaptic inhibition and their behavioral effects remain unclear. Here we report the existence of bidirectional alterations to the synaptic inhibition on CA1 pyramidal cells in the Ts2Cje mouse model of DS which are associated to impaired spatial memory. Furthermore, we identify triplication of the kainate receptor (KAR) encoding gene Grik1 as the cause of these phenotypes. Normalization of Grik1 dosage in Ts2Cje mice specifically restored spatial memory and reversed the bidirectional alterations to CA1 inhibition, but not the changes in synaptic plasticity or the other behavioral modifications observed. We propose that modified information gating caused by disturbed inhibitory tone rather than generalized overinhibition underlies some of the characteristic cognitive deficits in DS. Exaggerated synaptic inhibition is hypothesised to be a main cause of cognitive deficits in Down syndrome models. The authors identify triplication of the kainate receptor encoding gene Grik1 as the cause of memory deficits due to a reorganization of synaptic inhibition along the CA1 dendritic tree.
Collapse
|
30
|
Yu YE, Xing Z, Do C, Pao A, Lee EJ, Krinsky-McHale S, Silverman W, Schupf N, Tycko B. Genetic and epigenetic pathways in Down syndrome: Insights to the brain and immune system from humans and mouse models. PROGRESS IN BRAIN RESEARCH 2019; 251:1-28. [PMID: 32057305 PMCID: PMC7286740 DOI: 10.1016/bs.pbr.2019.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The presence of an extra copy of human chromosome 21 (Hsa21) leads to a constellation of phenotypic manifestations in Down syndrome (DS), including prominent effects on the brain and immune system. Intensive efforts to unravel the molecular mechanisms underlying these phenotypes may help developing effective therapies, both in DS and in the general population. Here we review recent progress in genetic and epigenetic analysis of trisomy 21 (Ts21). New mouse models of DS based on syntenic conservation of segments of the mouse and human chromosomes are starting to clarify the contributions of chromosomal subregions and orthologous genes to specific phenotypes in DS. The expression of genes on Hsa21 is regulated by epigenetic mechanisms, and with recent findings of highly recurrent gene-specific changes in DNA methylation patterns in brain and immune system cells with Ts21, the epigenomics of DS has become an active research area. Here we highlight the value of combining human studies with mouse models for defining DS critical genes and understanding the trans-acting effects of a simple chromosomal aneuploidy on genome-wide epigenetic patterning. These genetic and epigenetic studies are starting to uncover fundamental biological mechanisms, leading to insights that may soon become therapeutically relevant.
Collapse
Affiliation(s)
- Y Eugene Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States; Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, United States.
| | - Zhuo Xing
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Catherine Do
- Department of Biomedical Research, Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation and Hackensack-Meridian Health School of Medicine at Seton Hall University, Nutley, NJ, United States
| | - Annie Pao
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Eun Joon Lee
- Department of Biomedical Research, Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation and Hackensack-Meridian Health School of Medicine at Seton Hall University, Nutley, NJ, United States
| | - Sharon Krinsky-McHale
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Wayne Silverman
- Kennedy Krieger Institute and Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pediatrics, University of California at Irvine, Irvine, CA, United States
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| | - Benjamin Tycko
- Department of Biomedical Research, Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation and Hackensack-Meridian Health School of Medicine at Seton Hall University, Nutley, NJ, United States.
| |
Collapse
|
31
|
Muñiz Moreno MDM, Brault V, Birling MC, Pavlovic G, Herault Y. Modeling Down syndrome in animals from the early stage to the 4.0 models and next. PROGRESS IN BRAIN RESEARCH 2019; 251:91-143. [PMID: 32057313 DOI: 10.1016/bs.pbr.2019.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The genotype-phenotype relationship and the physiopathology of Down Syndrome (DS) have been explored in the last 20 years with more and more relevant mouse models. From the early age of transgenesis to the new CRISPR/CAS9-derived chromosomal engineering and the transchromosomic technologies, mouse models have been key to identify homologous genes or entire regions homologous to the human chromosome 21 that are necessary or sufficient to induce DS features, to investigate the complexity of the genetic interactions that are involved in DS and to explore therapeutic strategies. In this review we report the new developments made, how genomic data and new genetic tools have deeply changed our way of making models, extended our panel of animal models, and increased our understanding of the neurobiology of the disease. But even if we have made an incredible progress which promises to make DS a curable condition, we are facing new research challenges to nurture our knowledge of DS pathophysiology as a neurodevelopmental disorder with many comorbidities during ageing.
Collapse
Affiliation(s)
- Maria Del Mar Muñiz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Véronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Marie-Christine Birling
- Université de Strasbourg, CNRS, INSERM, PHENOMIN Institut Clinique de la Souris, Illkirch, France
| | - Guillaume Pavlovic
- Université de Strasbourg, CNRS, INSERM, PHENOMIN Institut Clinique de la Souris, Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Université de Strasbourg, CNRS, INSERM, PHENOMIN Institut Clinique de la Souris, Illkirch, France.
| |
Collapse
|
32
|
Lauterborn JC, Schultz MN, Le AA, Amani M, Friedman AE, Leach PT, Gall CM, Lynch GS, Crawley JN. Spaced training improves learning in Ts65Dn and Ube3a mouse models of intellectual disabilities. Transl Psychiatry 2019; 9:166. [PMID: 31182707 PMCID: PMC6557858 DOI: 10.1038/s41398-019-0495-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 12/29/2022] Open
Abstract
Benefits of distributed learning strategies have been extensively described in the human literature, but minimally investigated in intellectual disability syndromes. We tested the hypothesis that training trials spaced apart in time could improve learning in two distinct genetic mouse models of neurodevelopmental disorders characterized by intellectual impairments. As compared to training with massed trials, spaced training significantly improved learning in both the Ts65Dn trisomy mouse model of Down syndrome and the maternally inherited Ube3a mutant mouse model of Angelman syndrome. Spacing the training trials at 1 h intervals accelerated acquisition of three cognitive tasks by Ts65Dn mice: (1) object location memory, (2) novel object recognition, (3) water maze spatial learning. Further, (4) spaced training improved water maze spatial learning by Ube3a mice. In contrast, (5) cerebellar-mediated rotarod motor learning was not improved by spaced training. Corroborations in three assays, conducted in two model systems, replicated within and across two laboratories, confirm the strength of the findings. Our results indicate strong translational relevance of a behavioral intervention strategy for improving the standard of care in treating the learning difficulties that are characteristic and clinically intractable features of many neurodevelopmental disorders.
Collapse
Affiliation(s)
- J C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - M N Schultz
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - A A Le
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - M Amani
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - A E Friedman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Harvard University, Cambridge, MA, USA
| | - P T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Biogen Inc., Cambridge, MA, USA
| | - C M Gall
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - G S Lynch
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - J N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
33
|
Abstract
Down syndrome (DS; Trisomy 21) is the most common chromosomal disorder in humans. It has numerous associated neurologic phenotypes including intellectual disability, sleep apnea, seizures, behavioral problems, and dementia. With improved access to medical care, people with DS are living longer than ever before. As more individuals with DS reach old age, the necessity for further life span research is essential and cannot be overstated. There is currently a scarcity of information on common medical conditions encountered as individuals with DS progress into adulthood and old age. Conflicting information and uncertainty about the relative risk of dementia for adults with DS is a source of distress for the DS community that creates a major obstacle to proper evaluation and treatment. In this chapter, we discuss the salient neurologic phenotypes of DS, including Alzheimer's disease (AD), and current understanding of their biologic bases and management.
Collapse
Affiliation(s)
- Michael S Rafii
- Department of Neurology, Keck School of Medicine of the University of Southern California, San Diego, CA, United States
| | | | - Mariko Sawa
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
34
|
Developmental excitatory-to-inhibitory GABA polarity switch is delayed in Ts65Dn mice, a genetic model of Down syndrome. Neurobiol Dis 2018; 115:1-8. [PMID: 29550538 DOI: 10.1016/j.nbd.2018.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 03/01/2018] [Accepted: 03/07/2018] [Indexed: 11/23/2022] Open
Abstract
Down syndrome (DS) is the most frequent genetic cause of developmental abnormalities leading to intellectual disability. One notable phenomenon affecting the formation of nascent neural circuits during late developmental periods is developmental switch of GABA action from depolarizing to hyperpolarizing mode. We examined properties of this switch in DS using primary cultures and acute hippocampal slices from Ts65Dn mice, a genetic model of DS. Cultures of DIV3-DIV13 Ts65Dn and control normosomic (2 N) neurons were loaded with FURA-2 AM, and GABA action was assessed using local applications. In 2 N cultures, the number of GABA-activated cells dropped from ~100% to 20% between postnatal days 3-13 (P3-P13) reflecting the switch in GABA action polarity. In Ts65Dn cultures, the timing of this switch was delayed by 2-3 days. Next, microelectrode recordings of multi-unit activity (MUA) were performed in CA3 slices during bath application of the GABAA agonist isoguvacine. MUA frequency was increased in P8-P12 and reduced in P14-P22 slices reflecting the switch of GABA action from excitatory to inhibitory mode. The timing of this switch was delayed in Ts65Dn by approximately 2 days. Finally, frequency of giant depolarizing potentials (GDPs), a form of primordial neural activity, was significantly increased in slices from Ts65Dn pups at P12 and P14. These experimental evidences show that GABA action polarity switch is delayed in Ts65Dn model of DS, and that these changes lead to a delay in maturation of nascent neural circuits. These alterations may affect properties of neural circuits in adult animals and, therefore, represent a prospective target for pharmacotherapy of cognitive impairment in DS.
Collapse
|
35
|
Zorrilla de San Martin J, Delabar JM, Bacci A, Potier MC. GABAergic over-inhibition, a promising hypothesis for cognitive deficits in Down syndrome. Free Radic Biol Med 2018; 114:33-39. [PMID: 28993272 DOI: 10.1016/j.freeradbiomed.2017.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/01/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022]
Abstract
Down syndrome (DS), also known as trisomy 21, is the most common genetic cause of intellectual disability. It is also a model human disease for exploring consequences of gene dosage imbalance on complex phenotypes. Learning and memory impairments linked to intellectual disabilities in DS could result from synaptic plasticity deficits and excitatory-inhibitory alterations leading to changes in neuronal circuitry in the brain of affected individuals. Increasing number of studies in mouse and cellular models converge towards the assumption that excitatory-inhibitory imbalance occurs in DS, likely early during development. Thus increased inhibition appears to be a common trend that could explain synaptic and circuit disorganization. Interestingly using several potent pharmacological tools, preclinical studies strongly demonstrated that cognitive deficits could be restored in mouse models of DS. Clinical trials have not yet provided robust data for therapeutic application and additional studies are needed. Here we review the literature and our own published work emphasizing the over-inhibition hypothesis in DS and their links with gene dosage imbalance paving the way for future basic and clinical research.
Collapse
Affiliation(s)
- Javier Zorrilla de San Martin
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Jean-Maurice Delabar
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Alberto Bacci
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Marie-Claude Potier
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.
| |
Collapse
|
36
|
Stringer M, Goodlett CR, Roper RJ. Targeting trisomic treatments: optimizing Dyrk1a inhibition to improve Down syndrome deficits. Mol Genet Genomic Med 2017; 5:451-465. [PMID: 28944229 PMCID: PMC5606891 DOI: 10.1002/mgg3.334] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 12/11/2022] Open
Abstract
Overexpression of Dual-specificity tyrosine-phosphorylated regulated kinase 1A (DYRK1A), located on human chromosome 21, may alter molecular processes linked to developmental deficits in Down syndrome (DS). Trisomic DYRK1A is a rational therapeutic target, and although reductions in Dyrk1a genetic dosage have shown improvements in trisomic mouse models, attempts to reduce Dyrk1a activity by pharmacological mechanisms and correct these DS-associated phenotypes have been largely unsuccessful. Epigallocatechin-3-gallate (EGCG) inhibits DYRK1A activity in vitro and this action has been postulated to account for improvement of some DS-associated phenotypes that have been reported in preclinical studies and clinical trials. However, the beneficial effects of EGCG are inconsistent and there is no direct evidence that any observed improvement actually occurs through Dyrk1a inhibition. Inconclusive outcomes likely reflect a lack of knowledge about the tissue-specific patterns of spatial and temporal overexpression and elevated activity of Dyrk1a that may contribute to emerging DS traits during development. Emerging evidence indicates that Dyrk1a expression varies over the life span in DS mouse models, yet preclinical therapeutic treatments targeting Dyrk1a have largely not considered these developmental changes. Therapies intended to improve DS phenotypes through normalizing trisomic Dyrk1a need to optimize the timing and dose of treatment to match the spatiotemporal patterning of excessive Dyrk1a activity in relevant tissues. This will require more precise identification of developmental periods of vulnerability to enduring adverse effects of elevated Dyrk1a, representing the concurrence of increased Dyrk1a expression together with hypothesized tissue-specific-sensitive periods when Dyrk1a regulates cellular processes that shape the long-term functional properties of the tissue. Future efforts targeting inhibition of trisomic Dyrk1a should identify these putative spatiotemporally specific developmental sensitive periods and determine whether normalizing Dyrk1a activity then can lead to improved outcomes in DS phenotypes.
Collapse
Affiliation(s)
- Megan Stringer
- Department of PsychologyIUPUI402 North Blackford Street, LD 124IndianapolisIndiana46202-3275
| | - Charles R Goodlett
- Department of PsychologyIUPUI402 North Blackford Street, LD 124IndianapolisIndiana46202-3275
| | - Randall J Roper
- Department of BiologyIUPUI723 West Michigan Street SL 306IndianapolisIndiana46202-3275
| |
Collapse
|