1
|
Boeck B, Westmark CJ. Bibliometric Analysis and a Call for Increased Rigor in Citing Scientific Literature: Folic Acid Fortification and Neural Tube Defect Risk as an Example. Nutrients 2024; 16:2503. [PMID: 39125384 PMCID: PMC11313885 DOI: 10.3390/nu16152503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The health benefits of vitamin B9 (folate) are well documented, particularly in regard to neural tube defects during pregnancy; however, much remains to be learned regarding the health effects and risks of consuming folic acid supplements and foods fortified with folic acid. In 2020, our laboratory conducted a population-based analysis of the Food Fortification Initiative (FFI) dataset to determine the strength of the evidence regarding the prevalence of neural tube defects (NTD) at the national level in response to mandatory fortification of cereal grains with folic acid. We found a very weak correlation between the prevalence of NTDs and the level of folic acid fortification irrespective of the cereal grain fortified (wheat, maize, or rice). We found a strong linear relationship between reduced NTDs and higher socioeconomic status (SES). Our paper incited a debate on the proper statistics to employ for population-level data. Subsequently, there has been a large number of erroneous citations to our original work. The objective here was to conduct a bibliometric analysis to quantitate the accuracy of citations to Murphy and Westmark's publication entitled, "Folic Acid Fortification and Neural Tube Defect Risk: Analysis of the Food Fortification Initiative Dataset". We found a 70% inaccuracy rate. These findings highlight the dire need for increased rigor in citing scientific literature, particularly in regard to biomedical research that directly impacts public health policy.
Collapse
Affiliation(s)
- Brynne Boeck
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA;
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA;
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
2
|
Cahill CM, Sarang SS, Bakshi R, Xia N, Lahiri DK, Rogers JT. Neuroprotective Strategies and Cell-Based Biomarkers for Manganese-Induced Toxicity in Human Neuroblastoma (SH-SY5Y) Cells. Biomolecules 2024; 14:647. [PMID: 38927051 PMCID: PMC11201412 DOI: 10.3390/biom14060647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/24/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Manganese (Mn) is an essential heavy metal in the human body, while excess Mn leads to neurotoxicity, as observed in this study, where 100 µM of Mn was administered to the human neuroblastoma (SH-SY5Y) cell model of dopaminergic neurons in neurodegenerative diseases. We quantitated pathway and gene changes in homeostatic cell-based adaptations to Mn exposure. Utilizing the Gene Expression Omnibus, we accessed the GSE70845 dataset as a microarray of SH-SY5Y cells published by Gandhi et al. (2018) and applied statistical significance cutoffs at p < 0.05. We report 74 pathway and 10 gene changes with statistical significance. ReactomeGSA analyses demonstrated upregulation of histones (5 out of 10 induced genes) and histone deacetylases as a neuroprotective response to remodel/mitigate Mn-induced DNA/chromatin damage. Neurodegenerative-associated pathway changes occurred. NF-κB signaled protective responses via Sirtuin-1 to reduce neuroinflammation. Critically, Mn activated three pathways implicating deficits in purine metabolism. Therefore, we validated that urate, a purine and antioxidant, mitigated Mn-losses of viability in SH-SY5Y cells. We discuss Mn as a hypoxia mimetic and trans-activator of HIF-1α, the central trans-activator of vascular hypoxic mitochondrial dysfunction. Mn induced a 3-fold increase in mRNA levels for antioxidant metallothionein-III, which was induced 100-fold by hypoxia mimetics deferoxamine and zinc.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Sanjan S. Sarang
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Rachit Bakshi
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Ning Xia
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Debomoy K. Lahiri
- Department of Psychiatry and Medical & Molecular Genetics, Indiana Alzheimer’s Disease Research Center, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Jack T. Rogers
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| |
Collapse
|
3
|
Sokol DK, Lahiri DK. Alzheimer's drugs, APPlication for Down syndrome? Ageing Res Rev 2024; 96:102281. [PMID: 38513771 DOI: 10.1016/j.arr.2024.102281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Accumulation of the amyloid β (Aβ) peptide, derived from Aβ precursor protein (APP), is a trait of Down syndrome (DS), as is early development of dementia that resembles Alzheimer's disease (AD). Treatments for this AD in DS simply do not. New drug therapies for AD, e.g., Lecanemab, are monoclonal antibodies designed to clear amyloid plaques composed of Aβ. The increasingly real ability to target and dispose of Aβ favors the use of these drugs in individuals with AD in DS, and, perhaps as earlier intervention for cognitive impairment. We present pertinent similarities between DS and AD in adult DS subjects, discuss challenges to target APP metabolites, and suggest that recently developed antibody treatments against Aβ may be worth investigating to treat AD in DS.
Collapse
Affiliation(s)
- Deborah K Sokol
- Department of Neurology, Section Pediatrics, Indiana University of Medicine, Riley Hospital, 575 Riley Hospital Drive, Indianapolis, IN 46202, USA
| | - Debomoy K Lahiri
- Department of Psychiatry, 320 West 15th St, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, 320 West 15th St, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, 320 West 15th St, Indianapolis, IN 46202, USA; Indiana Alzheimer's Disease Research Center, 320 West 15th St, Indianapolis, IN 46202, USA.
| |
Collapse
|
4
|
Ma M, Yu Q, Delafield DG, Cui Y, Li Z, Li M, Wu W, Shi X, Westmark PR, Gutierrez A, Ma G, Gao A, Xu M, Xu W, Westmark CJ, Li L. On-Tissue Spatial Proteomics Integrating MALDI-MS Imaging with Shotgun Proteomics Reveals Soy Consumption-Induced Protein Changes in a Fragile X Syndrome Mouse Model. ACS Chem Neurosci 2024; 15:119-133. [PMID: 38109073 PMCID: PMC11127747 DOI: 10.1021/acschemneuro.3c00497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Fragile X syndrome (FXS), the leading cause of inherited intellectual disability and autism, is caused by the transcriptional silencing of the FMR1 gene, which encodes the fragile X messenger ribonucleoprotein (FMRP). FMRP interacts with numerous brain mRNAs that are involved in synaptic plasticity and implicated in autism spectrum disorders. Our published studies indicate that single-source, soy-based diets are associated with increased seizures and autism. Thus, there is an acute need for an unbiased protein marker identification in FXS in response to soy consumption. Herein, we present a spatial proteomics approach integrating mass spectrometry imaging with label-free proteomics in the FXS mouse model to map the spatial distribution and quantify levels of proteins in the hippocampus and hypothalamus brain regions. In total, 1250 unique peptides were spatially resolved, demonstrating the diverse array of peptidomes present in the tissue slices and the broad coverage of the strategy. A group of proteins that are known to be involved in glycolysis, synaptic transmission, and coexpression network analysis suggest a significant association between soy proteins and metabolic and synaptic processes in the Fmr1KO brain. Ultimately, this spatial proteomics work represents a crucial step toward identifying potential candidate protein markers and novel therapeutic targets for FXS.
Collapse
Affiliation(s)
- Min Ma
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Qinying Yu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Daniel G. Delafield
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Yusi Cui
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Zihui Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Miyang Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Wenxin Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Xudong Shi
- Division of Otolaryngology, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Pamela R. Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Alejandra Gutierrez
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Molecular Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Gui Ma
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Ang Gao
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Meng Xu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Molecular Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
5
|
Sokol DK, Lahiri DK. Neurodevelopmental disorders and microcephaly: how apoptosis, the cell cycle, tau and amyloid-β precursor protein APPly. Front Mol Neurosci 2023; 16:1201723. [PMID: 37808474 PMCID: PMC10556256 DOI: 10.3389/fnmol.2023.1201723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/08/2023] [Indexed: 10/10/2023] Open
Abstract
Recent studies promote new interest in the intersectionality between autism spectrum disorder (ASD) and Alzheimer's Disease. We have reported high levels of Amyloid-β Precursor Protein (APP) and secreted APP-alpha (sAPPa ) and low levels of amyloid-beta (Aβ) peptides 1-40 and 1-42 (Aβ40, Aβ42) in plasma and brain tissue from children with ASD. A higher incidence of microcephaly (head circumference less than the 3rd percentile) associates with ASD compared to head size in individuals with typical development. The role of Aβ peptides as contributors to acquired microcephaly in ASD is proposed. Aβ may lead to microcephaly via disruption of neurogenesis, elongation of the G1/S cell cycle, and arrested cell cycle promoting apoptosis. As the APP gene exists on Chromosome 21, excess Aβ peptides occur in Trisomy 21-T21 (Down's Syndrome). Microcephaly and some forms of ASD associate with T21, and therefore potential mechanisms underlying these associations will be examined in this review. Aβ peptides' role in other neurodevelopmental disorders that feature ASD and acquired microcephaly are reviewed, including dup 15q11.2-q13, Angelman and Rett syndrome.
Collapse
Affiliation(s)
- Deborah K. Sokol
- Section of Pediatrics, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Debomoy K. Lahiri
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
6
|
Sotgiu MA, Lo Jacono A, Barisano G, Saderi L, Cavassa V, Montella A, Crivelli P, Carta A, Sotgiu S. Brain perivascular spaces and autism: clinical and pathogenic implications from an innovative volumetric MRI study. Front Neurosci 2023; 17:1205489. [PMID: 37425010 PMCID: PMC10328421 DOI: 10.3389/fnins.2023.1205489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Our single-center case-control study aimed to evaluate the unclear glymphatic system alteration in autism spectrum disorder (ASD) through an innovative neuroimaging tool which allows to segment and quantify perivascular spaces in the white matter (WM-PVS) with filtering of non-structured noise and increase of the contrast-ratio between perivascular spaces and the surrounding parenchyma. Methods Briefly, files of 65 ASD and 71 control patients were studied. We considered: ASD type, diagnosis and severity level and comorbidities (i.e., intellectual disability, attention-deficit hyperactivity disorder, epilepsy, sleep disturbances). We also examined diagnoses other than ASD and their associated comorbidities in the control group. Results When males and females with ASD are included together, WM-PVS grade and WM-PVS volume do not significantly differ between the ASD group and the control group overall. We found, instead, that WM-PVS volume is significantly associated with male sex: males had higher WM-PVS volume compared to females (p = 0.01). WM-PVS dilation is also non-significantly associated with ASD severity and younger age (< 4 years). In ASD patients, higher WM-PVS volume was related with insomnia whereas no relation was found with epilepsy or IQ. Discussion We concluded that WM-PVS dilation can be a neuroimaging feature of male ASD patients, particularly the youngest and most severe ones, which may rely on male-specific risk factors acting early during neurodevelopment, such as a transient excess of extra-axial CSF volume. Our findings can corroborate the well-known strong male epidemiological preponderance of autism worldwide.
Collapse
Affiliation(s)
| | - Alessandro Lo Jacono
- Unit of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Giuseppe Barisano
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
| | - Laura Saderi
- Clinical Epidemiology and Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Vanna Cavassa
- Unit of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Paola Crivelli
- Radiology Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Alessandra Carta
- Unit of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Stefano Sotgiu
- Unit of Child Neuropsychiatry, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| |
Collapse
|
7
|
Frackowiak J, Mazur-Kolecka B. Intraneuronal accumulation of amyloid-β peptides as the pathomechanism linking autism and its co-morbidities: epilepsy and self-injurious behavior - the hypothesis. Front Mol Neurosci 2023; 16:1160967. [PMID: 37305553 PMCID: PMC10250631 DOI: 10.3389/fnmol.2023.1160967] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/28/2023] [Indexed: 06/13/2023] Open
Abstract
Autism spectrum disorder (ASD) is associated with enhanced processing of amyloid-β precursor protein (APP) by secretase-α, higher blood levels of sAPPα and intraneuronal accumulation of N-terminally truncated Aβ peptides in the brain cortex - mainly in the GABAergic neurons expressing parvalbumin - and subcortical structures. Brain Aβ accumulation has been also described in epilepsy-the frequent ASD co-morbidity. Furthermore, Aβ peptides have been shown to induce electroconvulsive episodes. Enhanced production and altered processing of APP, as well as accumulation of Aβ in the brain are also frequent consequences of traumatic brain injuries which result from self-injurious behaviors, another ASD co-morbidity. We discuss distinct consequences of accumulation of Aβ in the neurons and synapses depending on the Aβ species, their posttranslational modifications, concentration, level of aggregation and oligomerization, as well as brain structures, cell types and subcellular structures where it occurs. The biological effects of Aβ species which are discussed in the context of the pathomechanisms of ASD, epilepsy, and self-injurious behavior include modulation of transcription-both activation and repression; induction of oxidative stress; activation and alteration of membrane receptors' signaling; formation of calcium channels causing hyper-activation of neurons; reduction of GABAergic signaling - all of which lead to disruption of functions of synapses and neuronal networks. We conclude that ASD, epilepsy, and self-injurious behaviors all contribute to the enhanced production and accumulation of Aβ peptides which in turn cause and enhance dysfunctions of the neuronal networks that manifest as autism clinical symptoms, epilepsy, and self-injurious behaviors.
Collapse
|
8
|
Cencelli G, Pacini L, De Luca A, Messia I, Gentile A, Kang Y, Nobile V, Tabolacci E, Jin P, Farace MG, Bagni C. Age-Dependent Dysregulation of APP in Neuronal and Skin Cells from Fragile X Individuals. Cells 2023; 12:758. [PMID: 36899894 PMCID: PMC10000963 DOI: 10.3390/cells12050758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 03/04/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of monogenic intellectual disability and autism, caused by the absence of the functional fragile X messenger ribonucleoprotein 1 (FMRP). FXS features include increased and dysregulated protein synthesis, observed in both murine and human cells. Altered processing of the amyloid precursor protein (APP), consisting of an excess of soluble APPα (sAPPα), may contribute to this molecular phenotype in mice and human fibroblasts. Here we show an age-dependent dysregulation of APP processing in fibroblasts from FXS individuals, human neural precursor cells derived from induced pluripotent stem cells (iPSCs), and forebrain organoids. Moreover, FXS fibroblasts treated with a cell-permeable peptide that decreases the generation of sAPPα show restored levels of protein synthesis. Our findings suggest the possibility of using cell-based permeable peptides as a future therapeutic approach for FXS during a defined developmental window.
Collapse
Affiliation(s)
- Giulia Cencelli
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Laura Pacini
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Faculty of Medicine, UniCamillus, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Anastasia De Luca
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilenia Messia
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonietta Gentile
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, 00166 Rome, Italy
| | - Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Veronica Nobile
- Institute of Genomic Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Elisabetta Tabolacci
- Institute of Genomic Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria Giulia Farace
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
9
|
Westmark CJ, Filon MJ, Maina P, Steinberg LI, Ikonomidou C, Westmark PR. Effects of Soy-Based Infant Formula on Weight Gain and Neurodevelopment in an Autism Mouse Model. Cells 2022; 11:1350. [PMID: 35456030 PMCID: PMC9025435 DOI: 10.3390/cells11081350] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Mice fed soy-based diets exhibit increased weight gain compared to mice fed casein-based diets, and the effects are more pronounced in a model of fragile X syndrome (FXS; Fmr1KO). FXS is a neurodevelopmental disability characterized by intellectual impairment, seizures, autistic behavior, anxiety, and obesity. Here, we analyzed body weight as a function of mouse age, diet, and genotype to determine the effect of diet (soy, casein, and grain-based) on weight gain. We also assessed plasma protein biomarker expression and behavior in response to diet. Juvenile Fmr1KO mice fed a soy protein-based rodent chow throughout gestation and postnatal development exhibit increased weight gain compared to mice fed a casein-based purified ingredient diet or grain-based, low phytoestrogen chow. Adolescent and adult Fmr1KO mice fed a soy-based infant formula diet exhibited increased weight gain compared to reference diets. Increased body mass was due to increased lean mass. Wild-type male mice fed soy-based infant formula exhibited increased learning in a passive avoidance paradigm, and Fmr1KO male mice had a deficit in nest building. Thus, at the systems level, consumption of soy-based diets increases weight gain and affects behavior. At the molecular level, a soy-based infant formula diet was associated with altered expression of numerous plasma proteins, including the adipose hormone leptin and the β-amyloid degrading enzyme neprilysin. In conclusion, single-source, soy-based diets may contribute to the development of obesity and the exacerbation of neurological phenotypes in developmental disabilities, such as FXS.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| | - Mikolaj J. Filon
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Undergraduate Research Program, University of Wisconsin, Madison, WI 53706, USA
| | - Patricia Maina
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Molecular Environmental Toxicology Summer Research Opportunities Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren I. Steinberg
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Undergraduate Research Program, University of Wisconsin, Madison, WI 53706, USA
| | - Chrysanthy Ikonomidou
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
| | - Pamela R. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
| |
Collapse
|
10
|
Siewert-Rocks KM, Kim SS, Yao DW, Shi H, Price AL. Leveraging gene co-regulation to identify gene sets enriched for disease heritability. Am J Hum Genet 2022; 109:393-404. [PMID: 35108496 PMCID: PMC8948163 DOI: 10.1016/j.ajhg.2022.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022] Open
Abstract
Identifying gene sets that are associated to disease can provide valuable biological knowledge, but a fundamental challenge of gene set analyses of GWAS data is linking disease-associated SNPs to genes. Transcriptome-wide association studies (TWASs) detect associations between the genetically predicted expression of a gene and disease risk, thus implicating candidate disease genes. However, causal disease genes at TWAS-associated loci generally remain unknown due to gene co-regulation, which leads to correlations across genes in predicted expression. We developed a method, gene co-regulation score (GCSC) regression, to identify gene sets that are enriched for disease heritability explained by predicted expression. GCSC regresses TWAS chi-square statistics on gene co-regulation scores reflecting correlations in predicted gene expression; a gene set is enriched for heritability if genes with high co-regulation to the set have higher TWAS chi-square statistics than genes with low co-regulation to the set, beyond what is expected based on co-regulation to all genes. We verified via simulations that GCSC is well calibrated and well powered. We applied GCSC to gene expression data from GTEx (48 tissues) and GWAS summary statistics for 43 independent diseases and complex traits analyzing a broad set of biological pathways and specifically expressed gene sets. We identified many enriched sets, recapitulating known biology. For Alzheimer disease, we detected evidence of an immune basis, and specifically a role for antigen presentation, in analyses of both biological pathways and specifically expressed gene sets. Our results highlight the advantages of leveraging gene co-regulation within the TWAS framework to identify enriched gene sets.
Collapse
Affiliation(s)
- Katherine M Siewert-Rocks
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Samuel S Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Douglas W Yao
- Program in Systems, Synthetic, and Quantitative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Huwenbo Shi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Wang R, Chopra N, Nho K, Maloney B, Obukhov AG, Nelson PT, Counts SE, Lahiri DK. Human microRNA (miR-20b-5p) modulates Alzheimer's disease pathways and neuronal function, and a specific polymorphism close to the MIR20B gene influences Alzheimer's biomarkers. Mol Psychiatry 2022; 27:1256-1273. [PMID: 35087196 PMCID: PMC9054681 DOI: 10.1038/s41380-021-01351-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with loss of cognitive, executive, and other mental functions, and is the most common form of age-related dementia. Amyloid-β peptide (Aβ) contributes to the etiology and progression of the disease. Aβ is derived from the amyloid-β precursor protein (APP). Multiple microRNA (miRNA) species are also implicated in AD. We report that human hsa-miR20b-5p (miR-20b), produced from the MIR20B gene on Chromosome X, may play complex roles in AD pathogenesis, including Aβ regulation. Specifically, miR-20b-5p miRNA levels were altered in association with disease progression in three regions of the human brain: temporal neocortex, cerebellum, and posterior cingulate cortex. In cultured human neuronal cells, miR-20b-5p treatment interfered with calcium homeostasis, neurite outgrowth, and branchpoints. A single-nucleotide polymorphism (SNP) upstream of the MIR20B gene (rs13897515) associated with differences in levels of cerebrospinal fluid (CSF) Aβ1-42 and thickness of the entorhinal cortex. We located a miR-20b-5p binding site in the APP mRNA 3'-untranslated region (UTR), and treatment with miR-20b-5p reduced APP mRNA and protein levels. Network analysis of protein-protein interactions and gene coexpression revealed other important potential miR-20b-5p targets among AD-related proteins/genes. MiR-20b-5p, a miRNA that downregulated APP, was paradoxically associated with an increased risk for AD. However, miR-20b-5p also reduced, and the blockade of APP by siRNA likewise reduced calcium influx. As APP plays vital roles in neuronal health and does not exist solely to be the source of "pathogenic" Aβ, the molecular etiology of AD is likely to not just be a disease of "excess" but a disruption of delicate homeostasis.
Collapse
Affiliation(s)
- Ruizhi Wang
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nipun Chopra
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- DePauw University, Greencastle, IN, 46135, USA
| | - Kwangsik Nho
- Radiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bryan Maloney
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexander G Obukhov
- Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Kentucky Alzheimer's Disease Research Center, Lexington, KY, 40536, USA
| | - Scott E Counts
- Departments of Translational Neuroscience & Family Medicine, Michigan State University, Grand Rapids, and Michigan Alzheimer's Disease Research Center, Ann Arbor, MI, USA
| | - Debomoy K Lahiri
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
12
|
Nadeem MS, Hosawi S, Alshehri S, Ghoneim MM, Imam SS, Murtaza BN, Kazmi I. Symptomatic, Genetic, and Mechanistic Overlaps between Autism and Alzheimer's Disease. Biomolecules 2021; 11:1635. [PMID: 34827633 PMCID: PMC8615882 DOI: 10.3390/biom11111635] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023] Open
Abstract
Autism spectrum disorder (ASD) and Alzheimer's disease (AD) are neurodevelopmental and neurodegenerative disorders affecting two opposite ends of life span, i.e., childhood and old age. Both disorders pose a cumulative threat to human health, with the rate of incidences increasing considerably worldwide. In the context of recent developments, we aimed to review correlated symptoms and genetics, and overlapping aspects in the mechanisms of the pathogenesis of ASD and AD. Dementia, insomnia, and weak neuromuscular interaction, as well as communicative and cognitive impairments, are shared symptoms. A number of genes and proteins linked with both disorders have been tabulated, including MECP2, ADNP, SCN2A, NLGN, SHANK, PTEN, RELN, and FMR1. Theories about the role of neuron development, processing, connectivity, and levels of neurotransmitters in both disorders have been discussed. Based on the recent literature, the roles of FMRP (Fragile X mental retardation protein), hnRNPC (heterogeneous ribonucleoprotein-C), IRP (Iron regulatory proteins), miRNAs (MicroRNAs), and α-, β0, and γ-secretases in the posttranscriptional regulation of cellular synthesis and processing of APP (amyloid-β precursor protein) have been elaborated to describe the parallel and overlapping routes and mechanisms of ASD and AD pathogenesis. However, the interactive role of genetic and environmental factors, oxidative and metal ion stress, mutations in the associated genes, and alterations in the related cellular pathways in the development of ASD and AD needs further investigation.
Collapse
Affiliation(s)
- Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology (AUST), Abbottabad 22310, Pakistan;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| |
Collapse
|
13
|
Griesi-Oliveira K, Fogo MS, Pinto BGG, Alves AY, Suzuki AM, Morales AG, Ezquina S, Sosa OJ, Sutton GJ, Sunaga-Franze DY, Bueno AP, Seabra G, Sardinha L, Costa SS, Rosenberg C, Zachi EC, Sertie AL, Martins-de-Souza D, Reis EM, Voineagu I, Passos-Bueno MR. Transcriptome of iPSC-derived neuronal cells reveals a module of co-expressed genes consistently associated with autism spectrum disorder. Mol Psychiatry 2021; 26:1589-1605. [PMID: 32060413 PMCID: PMC8159745 DOI: 10.1038/s41380-020-0669-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/11/2019] [Accepted: 01/28/2020] [Indexed: 01/02/2023]
Abstract
Evaluation of expression profile in autism spectrum disorder (ASD) patients is an important approach to understand possible similar functional consequences that may underlie disease pathophysiology regardless of its genetic heterogeneity. Induced pluripotent stem cell (iPSC)-derived neuronal models have been useful to explore this question, but larger cohorts and different ASD endophenotypes still need to be investigated. Moreover, whether changes seen in this in vitro model reflect previous findings in ASD postmortem brains and how consistent they are across the studies remain underexplored questions. We examined the transcriptome of iPSC-derived neuronal cells from a normocephalic ASD cohort composed mostly of high-functioning individuals and from non-ASD individuals. ASD patients presented expression dysregulation of a module of co-expressed genes involved in protein synthesis in neuronal progenitor cells (NPC), and a module of genes related to synapse/neurotransmission and a module related to translation in neurons. Proteomic analysis in NPC revealed potential molecular links between the modules dysregulated in NPC and in neurons. Remarkably, the comparison of our results to a series of transcriptome studies revealed that the module related to synapse has been consistently found as upregulated in iPSC-derived neurons-which has an expression profile more closely related to fetal brain-while downregulated in postmortem brain tissue, indicating a reliable association of this network to the disease and suggesting that its dysregulation might occur in different directions across development in ASD individuals. Therefore, the expression pattern of this network might be used as biomarker for ASD and should be experimentally explored as a therapeutic target.
Collapse
Affiliation(s)
- K Griesi-Oliveira
- Hospital Israelita Albert Einstein, São Paulo, Brazil.
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| | - M S Fogo
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - B G G Pinto
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - A Y Alves
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - A M Suzuki
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - A G Morales
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - S Ezquina
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - O J Sosa
- Programa Interunidades de Pós-Graduação em Bioinformática, Universidade de São Paulo, São Paulo, Brazil
| | - G J Sutton
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - D Y Sunaga-Franze
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - A P Bueno
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - G Seabra
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - L Sardinha
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - S S Costa
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - C Rosenberg
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - E C Zachi
- Núcleo de Neurociências e Comportamento, Departamento de Psicologia Experimental, Instituto de Psicologia, Universidade de São Paulo, São Paulo, Brazil
| | - A L Sertie
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - D Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), São Paulo, Brazil
- Experimental Medicine Research Cluster (EMC), University of Campinas, Campinas, Brazil
| | - E M Reis
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - I Voineagu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - M R Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
14
|
Lahiri DK, Maloney B, Wang R, Sokol DK, Rogers JT, Westmark CJ. How autism and Alzheimer's disease are TrAPPed. Mol Psychiatry 2021; 26:26-29. [PMID: 33184495 PMCID: PMC9487718 DOI: 10.1038/s41380-020-00928-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/03/2020] [Accepted: 10/19/2020] [Indexed: 02/05/2023]
Affiliation(s)
- Debomoy K Lahiri
- Department of Psychiatry and Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Bryan Maloney
- Department of Psychiatry and Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ruizhi Wang
- Department of Psychiatry and Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Deborah K Sokol
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jack T Rogers
- Department of Psychiatry, Neurochemistry lab, MGH, Harvard Medical School, Charlestown, MA, USA
| | - Cara J Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
15
|
Jęśko H, Cieślik M, Gromadzka G, Adamczyk A. Dysfunctional proteins in neuropsychiatric disorders: From neurodegeneration to autism spectrum disorders. Neurochem Int 2020; 141:104853. [PMID: 32980494 DOI: 10.1016/j.neuint.2020.104853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Despite fundamental differences in disease course and outcomes, neurodevelopmental (autism spectrum disorders - ASD) and neurodegenerative disorders (Alzheimer's disease - AD and Parkinson's disease - PD) present surprising, common traits in their molecular pathomechanisms. Uncontrolled oligomerization and aggregation of amyloid β (Aβ), microtubule-associated protein (MAP) tau, or α-synuclein (α-syn) contribute to synaptic impairment and the ensuing neuronal death in both AD and PD. Likewise, the pathogenesis of ASD may be attributed, at least in part, to synaptic dysfunction; attention has also been recently paid to irregularities in the metabolism and function of the Aβ precursor protein (APP), tau, or α-syn. Commonly affected elements include signaling pathways that regulate cellular metabolism and survival such as insulin/insulin-like growth factor (IGF) - PI3 kinase - Akt - mammalian target of rapamycin (mTOR), and a number of key synaptic proteins critically involved in neuronal communication. Understanding how these shared pathomechanism elements operate in different conditions may help identify common targets and therapeutic approaches.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Magdalena Cieślik
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Grażyna Gromadzka
- Cardinal Stefan Wyszynski University, Faculty of Medicine. Collegium Medicum, Wóycickiego 1/3, 01-938, Warsaw, Poland.
| | - Agata Adamczyk
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| |
Collapse
|
16
|
Westmark CJ, Maloney B, Alisch RS, Sokol DK, Lahiri DK. FMRP Regulates the Nuclear Export of Adam9 and Psen1 mRNAs: Secondary Analysis of an N 6-Methyladenosine Dataset. Sci Rep 2020; 10:10781. [PMID: 32612155 PMCID: PMC7329887 DOI: 10.1038/s41598-020-66394-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/19/2020] [Indexed: 01/19/2023] Open
Abstract
Fragile X mental retardation protein (FMRP) binds to and regulates the translation of amyloid-β protein precursor (App) mRNA, but the detailed mechanism remains to be determined. Differential methylation of App mRNA could underlie FMRP binding, message localization and translation efficiency. We sought to determine the role of FMRP and N6-methyladeonsine (m6A) on nuclear export of App mRNA. We utilized the m6A dataset by Hsu and colleagues to identify m6A sites in App mRNA and to determine if the abundance of message in the cytoplasm relative to the nucleus is altered in Fmr1 knockout mouse brain cortex. Given that processing of APP to Aβ and soluble APP alpha (sAPPα) contributes to disease phenotypes, we also investigated whether Fmr1KO associates with nuclear export of the mRNAs for APP protein processing enzymes, including β-site amyloid cleaving enzyme (Bace1), A disintegrin and metalloproteinases (Adams), and presenilins (Psen). Fmr1KO did not alter the nuclear/cytoplasmic abundance of App mRNA. Of 36 validated FMRP targets, 35 messages contained m6A peaks but only Agap2 mRNA was selectively enriched in Fmr1KO nucleus. The abundance of the APP processing enzymes Adam9 and Psen1 mRNA, which code for a minor alpha-secretase and gamma-secretase, respectively, were selectively enriched in wild type cytoplasm.
Collapse
Affiliation(s)
- Cara J Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Bryan Maloney
- Department of Psychiatry, Indiana Alzheimer Disease Center, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Deborah K Sokol
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K Lahiri
- Department of Psychiatry, Indiana Alzheimer Disease Center, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
17
|
Transient upregulation of translational efficiency in prodromal and early symptomatic Tg2576 mice contributes to Aβ pathology. Neurobiol Dis 2020; 139:104787. [PMID: 32032729 DOI: 10.1016/j.nbd.2020.104787] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/14/2020] [Accepted: 01/31/2020] [Indexed: 12/20/2022] Open
Abstract
TG2576 mice show highest levels of the full length mutant Swedish Human Amyloid Precursor Protein (APPKM670/671LN) during prodromal and early sympotomatic stages. Interestingly, this occurs in association with the unbalanced expression of two of its RNA Binding proteins (RBPs) opposite regulators, the Fragile-X Mental Retardation Protein (FMRP) and the heteronuclear Ribonucleoprotein C (hnRNP C). Whether an augmentation in overall translational efficiency also contributes to the elevation of APP levels at those early developmental stages is currently unknown. We investigated this possibility by performing a longitudinal polyribosome profiling analysis of APP mRNA and protein in total hippocampal extracts from Tg2576 mice. Results showed that protein polysomal signals were exclusively detected in pre-symptomatic (1 months) and early symptomatic (3 months) mutant mice. Differently, hAPP mRNA polysomal signals were detected at any age, but a peak of expression was found when mice were 3-month old. Consistent with an early but transient rise of translational efficiency, the phosphorylated form of the initial translation factor eIF2α (p-eIF2α) was reduced at pre-symptomatic and early symptomatic stages, whereas it was increased at the fully symptomatic stage. Pharmacological downregulation of overall translation in early symptomatic mutants was then found to reduce hippocampal levels of full length APP, Aβspecies, BACE1 and Caspase-3, to rescue predominant LTD at hippocampal synapses, to revert dendritic spine loss and memory alterations, and to reinstate memory-induced c-fosactivation. Altogether, our findings demonstrate that overall translation is upregulated in prodromal and early symptomatic Tg2576 mice, and that restoring proper translational control at the onset of AD-like symptoms blocks the emergence of the AD-like phenotype.
Collapse
|
18
|
McLane RD, Schmitt LM, Pedapati EV, Shaffer RC, Dominick KC, Horn PS, Gross C, Erickson CA. Peripheral Amyloid Precursor Protein Derivative Expression in Fragile X Syndrome. Front Integr Neurosci 2019; 13:49. [PMID: 31551722 PMCID: PMC6733993 DOI: 10.3389/fnint.2019.00049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/16/2019] [Indexed: 01/08/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and is associated with increased risk for autism spectrum disorder (ASD), anxiety, ADHD, and epilepsy. While our understanding of FXS pathophysiology has improved, a lack of validated blood-based biomarkers of disease continues to impede bench-to-bedside efforts. To meet this demand, there is a growing effort to discover a reliable biomarker to inform treatment discovery and evaluate treatment target engagement. Such a marker, amyloid-beta precursor protein (APP), has shown potential dysregulation in the absence of fragile X mental retardation protein (FMRP) and may therefore be associated with FXS pathophysiology. While APP is best understood in the context of Alzheimer disease, there is a growing body of evidence suggesting the molecule and its derivatives play a broader role in regulating neuronal hyperexcitability, a well-characterized phenotype in FXS. To evaluate the viability of APP as a peripheral biological marker in FXS, we conducted an exploratory ELISA-based evaluation of plasma APP-related species involving 27 persons with FXS (mean age: 22.0 ± 11.5) and 25 age- and sex-matched persons with neurotypical development (mean age: 21.1 ± 10.7). Peripheral levels of both Aβ(1–40) and Aβ(1–42) were increased, while sAPPα was significantly decreased in persons with FXS as compared to control participants. These results suggest that dysregulated APP processing, with potential preferential β-secretase processing, may be a readily accessible marker of FXS pathophysiology.
Collapse
Affiliation(s)
- Richard D McLane
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Lauren M Schmitt
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ernest V Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Rebecca C Shaffer
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kelli C Dominick
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Paul S Horn
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
19
|
Pain O, Pocklington AJ, Holmans PA, Bray NJ, O’Brien HE, Hall LS, Pardiñas AF, O’Donovan MC, Owen MJ, Anney R. Novel Insight Into the Etiology of Autism Spectrum Disorder Gained by Integrating Expression Data With Genome-wide Association Statistics. Biol Psychiatry 2019; 86:265-273. [PMID: 31230729 PMCID: PMC6664597 DOI: 10.1016/j.biopsych.2019.04.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND A recent genome-wide association study (GWAS) of autism spectrum disorder (ASD) (ncases = 18,381, ncontrols = 27,969) has provided novel opportunities for investigating the etiology of ASD. Here, we integrate the ASD GWAS summary statistics with summary-level gene expression data to infer differential gene expression in ASD, an approach called transcriptome-wide association study (TWAS). METHODS Using FUSION software, ASD GWAS summary statistics were integrated with predictors of gene expression from 16 human datasets, including adult and fetal brains. A novel adaptation of established statistical methods was then used to test for enrichment within candidate pathways and specific tissues and at different stages of brain development. The proportion of ASD heritability explained by predicted expression of genes in the TWAS was estimated using stratified linkage disequilibrium score regression. RESULTS This study identified 14 genes as significantly differentially expressed in ASD, 13 of which were outside of known genome-wide significant loci (±500 kb). XRN2, a gene proximal to an ASD GWAS locus, was inferred to be significantly upregulated in ASD, providing insight into the functional consequence of this associated locus. One novel transcriptome-wide significant association from this study is the downregulation of PDIA6, which showed minimal evidence of association in the GWAS, and in gene-based analysis using MAGMA. Predicted gene expression in this study accounted for 13.0% of the total ASD single nucleotide polymorphism heritability. CONCLUSIONS This study has implicated several genes as significantly up/downregulated in ASD, providing novel and useful information for subsequent functional studies. This study also explores the utility of TWAS-based enrichment analysis and compares TWAS results with a functionally agnostic approach.
Collapse
Affiliation(s)
- Oliver Pain
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Andrew J. Pocklington
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Peter A. Holmans
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Nicholas J. Bray
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Heath E. O’Brien
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Lynsey S. Hall
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Antonio F. Pardiñas
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael C. O’Donovan
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael J. Owen
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Richard Anney
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
20
|
Westmark CJ. Fragile X and APP: a Decade in Review, a Vision for the Future. Mol Neurobiol 2019; 56:3904-3921. [PMID: 30225775 PMCID: PMC6421119 DOI: 10.1007/s12035-018-1344-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/05/2018] [Indexed: 10/28/2022]
Abstract
Fragile X syndrome (FXS) is a devastating developmental disability that has profound effects on cognition, behavior, and seizure susceptibility. There are currently no treatments that target the underlying cause of the disorder, and recent clinical trials have been unsuccessful. In 2007, seminal work demonstrated that amyloid-beta protein precursor (APP) is dysregulated in Fmr1KO mice through a metabotropic glutamate receptor 5 (mGluR5)-dependent pathway. These findings raise the hypotheses that: (1) APP and/or APP metabolites are potential therapeutic targets as well as biomarkers for FXS and (2) mGluR5 inhibitors may be beneficial in the treatment of Alzheimer's disease. Herein, advances in the field over the past decade that have reproduced and greatly expanded upon these original findings are reviewed, and required experimentation to validate APP metabolites as potential disease biomarkers as well as therapeutic targets for FXS are discussed.
Collapse
Affiliation(s)
- Cara J Westmark
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, Room 3619, 1300 University Avenue, Madison, WI, USA.
| |
Collapse
|
21
|
Sokol DK, Maloney B, Westmark CJ, Lahiri DK. Novel Contribution of Secreted Amyloid-β Precursor Protein to White Matter Brain Enlargement in Autism Spectrum Disorder. Front Psychiatry 2019; 10:165. [PMID: 31024350 PMCID: PMC6469489 DOI: 10.3389/fpsyt.2019.00165] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/06/2019] [Indexed: 12/27/2022] Open
Abstract
The most replicated neuroanatomical finding in autism is the tendency toward brain overgrowth, especially in younger children. Research shows that both gray and white matter are enlarged. Proposed mechanisms underlying brain enlargement include abnormal inflammatory and neurotrophic signals that lead to excessive, aberrant dendritic connectivity via disrupted pruning and cell adhesion, and enlargement of white matter due to excessive gliogenesis and increased myelination. Amyloid-β protein precursor (βAPP) and its metabolites, more commonly associated with Alzheimer's disease (AD), are also dysregulated in autism plasma and brain tissue samples. This review highlights findings that demonstrate how one βAPP metabolite, secreted APPα, and the ADAM family α-secretases, may lead to increased brain matter, with emphasis on increased white matter as seen in autism. sAPPα and the ADAM family α-secretases contribute to the anabolic, non-amyloidogenic pathway, which is in contrast to the amyloid (catabolic) pathway known to contribute to Alzheimer disease. The non-amyloidogenic pathway could produce brain enlargement via genetic mechanisms affecting mRNA translation and polygenic factors that converge on molecular pathways (mitogen-activated protein kinase/MAPK and mechanistic target of rapamycin/mTOR), promoting neuroinflammation. A novel mechanism linking the non-amyloidogenic pathway to white matter enlargement is proposed: α-secretase and/or sAPPα, activated by ERK receptor signaling activates P13K/AKt/mTOR and then Rho GTPases favoring myelination via oligodendrocyte progenitor cell (OPC) activation of cofilin. Applying known pathways in AD to autism should allow further understanding and provide options for new drug targets.
Collapse
Affiliation(s)
- Deborah K. Sokol
- Pediatrics Section, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bryan Maloney
- Indiana Alzheimers Disease Center, Department of Psychiatry, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
| | - Debomoy K. Lahiri
- Indiana Alzheimers Disease Center, Department of Psychiatry, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
22
|
Abraham JR, Barnard J, Wang H, Noritz GH, Yeganeh M, Buhas D, Natowicz MR. Proteomic investigations of human HERC2 mutants: Insights into the pathobiology of a neurodevelopmental disorder. Biochem Biophys Res Commun 2019; 512:421-427. [PMID: 30902390 DOI: 10.1016/j.bbrc.2019.02.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/27/2019] [Indexed: 01/11/2023]
Abstract
HERC2 is a giant protein with E3 ubiquitin ligase activity and other known and suspected functions. Mutations of HERC2 are implicated in the pathogenesis of various cancers and result in severe neurological conditions in Herc2-mutant mice. Recently, a pleotropic autosomal recessive HERC2-associated syndrome of intellectual disability, autism and variable neurological deficits was described; its pathogenetic basis is largely unknown. Using peripheral blood-derived lymphoblasts from 3 persons with homozygous HERC2 variants and 14 age- and gender-matched controls, we performed label-free unbiased HPLC-tandem mass spectrometry-based proteomic analyses to provide insights into HERC2-mediated pathobiology. We found that out of 3427 detected proteins, there were 812 differentially expressed proteins between HERC2-cases vs. controls. 184 canonical pathways were enriched after FDR adjustment, including mitochondrial function, energy metabolism, EIF2 signaling, immune functions, ubiquitination and DNA repair. Ingenuity Pathway Analysis® identified 209 upstream regulators that could drive the differential expression, prominent amongst which were neurodegeneration-associated proteins. Differentially expressed protein interaction networks highlighted themes of immune function/dysfunction, regulation of cell cycle/cell death, and energy metabolism. Overall, the analysis of the HERC2-associated proteome revealed striking differential protein expression between cases and controls. The large number of differentially expressed proteins likely reflects HERC2's multiple domains and numerous interacting proteins. Our canonical pathway and protein interaction network findings suggest derangements of multiple pathways in HERC2-associated disease.
Collapse
Affiliation(s)
- Joseph R Abraham
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Heng Wang
- DDC Clinic, Center for Special Needs Children, Middlefield, OH, USA
| | - Garey H Noritz
- Complex Health Care Program, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mehdi Yeganeh
- Department of Medical Genetics, McGill University Health Centre, Montreal, Canada
| | - Daniela Buhas
- Department of Medical Genetics, McGill University Health Centre, Montreal, Canada
| | - Marvin R Natowicz
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA; Pathology and Laboratory Medicine, Genomic Medicine, Neurological and Pediatrics Institutes, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
23
|
Carter CJ. Autism genes and the leukocyte transcriptome in autistic toddlers relate to pathogen interactomes, infection and the immune system. A role for excess neurotrophic sAPPα and reduced antimicrobial Aβ. Neurochem Int 2019; 126:36-58. [PMID: 30862493 DOI: 10.1016/j.neuint.2019.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Prenatal and early childhood infections have been implicated in autism. Many autism susceptibility genes (206 Autworks genes) are localised in the immune system and are related to immune/infection pathways. They are enriched in the host/pathogen interactomes of 18 separate microbes (bacteria/viruses and fungi) and to the genes regulated by bacterial toxins, mycotoxins and Toll-like receptor ligands. This enrichment was also observed for misregulated genes from a microarray study of leukocytes from autistic toddlers. The upregulated genes from this leukocyte study also matched the expression profiles in response to numerous infectious agents from the Broad Institute molecular signatures database. They also matched genes related to sudden infant death syndrome and autism comorbid conditions (autoimmune disease, systemic lupus erythematosus, diabetes, epilepsy and cardiomyopathy) as well as to estrogen and thyrotropin responses and to those upregulated by different types of stressors including oxidative stress, hypoxia, endoplasmic reticulum stress, ultraviolet radiation or 2,4-dinitrofluorobenzene, a hapten used to develop allergic skin reactions in animal models. The oxidative/integrated stress response is also upregulated in the autism brain and may contribute to myelination problems. There was also a marked similarity between the expression signatures of autism and Alzheimer's disease, and 44 shared autism/Alzheimer's disease genes are almost exclusively expressed in the blood-brain barrier. However, in contrast to Alzheimer's disease, levels of the antimicrobial peptide beta-amyloid are decreased and the levels of the neurotrophic/myelinotrophic soluble APP alpha are increased in autism, together with an increased activity of α-secretase. sAPPα induces an increase in glutamatergic and a decrease in GABA-ergic synapses creating and excitatory/inhibitory imbalance that has also been observed in autism. A literature survey showed that multiple autism genes converge on APP processing and that many are able to increase sAPPalpha at the expense of beta-amyloid production. A genetically programmed tilt of this axis towards an overproduction of neurotrophic/gliotrophic sAPPalpha and underproduction of antimicrobial beta-amyloid may explain the brain overgrowth and myelination dysfunction, as well as the involvement of pathogens in autism.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, 41C Marina, Saint Leonard's on Sea, TN38 0BU, East Sussex, UK.
| |
Collapse
|
24
|
Maurin T, Bardoni B. Fragile X Mental Retardation Protein: To Be or Not to Be a Translational Enhancer. Front Mol Biosci 2018; 5:113. [PMID: 30619879 PMCID: PMC6297276 DOI: 10.3389/fmolb.2018.00113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Thomas Maurin
- Université Côte d'Azur, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, Valbonne, France.,CNRS LIA "Neogenex", Valbonne, France
| | - Barbara Bardoni
- CNRS LIA "Neogenex", Valbonne, France.,Université Côte d'Azur, INSERM, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, Valbonne, France
| |
Collapse
|
25
|
Grayaa S, Zerbinati C, Messedi M, HadjKacem I, Chtourou M, Ben Touhemi D, Naifar M, Ayadi H, Ayedi F, Iuliano L. Plasma oxysterol profiling in children reveals 24-hydroxycholesterol as a potential marker for Autism Spectrum Disorders. Biochimie 2018; 153:80-85. [DOI: 10.1016/j.biochi.2018.04.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/29/2018] [Indexed: 12/19/2022]
|
26
|
Westmark PR, Dekundy A, Gravius A, Danysz W, Westmark CJ. Rescue of Fmr1 KO phenotypes with mGluR 5 inhibitors: MRZ-8456 versus AFQ-056. Neurobiol Dis 2018; 119:190-198. [PMID: 30125640 DOI: 10.1016/j.nbd.2018.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 12/23/2022] Open
Abstract
Metabotropic glutamate receptor 5 (mGluR5) is a drug target for central nervous system disorders such as fragile X syndrome that involve excessive glutamate-induced excitation. We tested the efficacy of a novel negative allosteric modulator of mGluR5 developed by Merz Pharmaceuticals, MRZ-8456, in comparison to MPEP and AFQ-056 (Novartis, a.k.a. mavoglurant) in both in vivo and in vitro assays in a mouse model of fragile X syndrome, Fmr1KO mice. The in vivo assays included susceptibility to audiogenic-induced seizures and pharmacokinetic measurements of drug availability. The in vitro assays included dose response assessments of biomarker expression and dendritic spine length and density in cultured primary neurons. Both MRZ-8456 and AFQ-056 attenuated wild running and audiogenic-induced seizures in Fmr1KO mice with similar pharmacokinetic profiles. Both drugs significantly reduced dendritic expression of amyloid-beta protein precursor (APP) and rescued the ratio of mature to immature dendritic spines. These findings demonstrate that MRZ-8456, a drug being developed for the treatment of motor complications of L-DOPA in Parkinson's disease and which completed a phase I clinical trial, is effective in attenuating both well-established (seizures and dendritic spine maturity) and exploratory biomarker (APP expression) phenotypes in a mouse model of fragile X syndrome.
Collapse
Affiliation(s)
- Pamela R Westmark
- University of Wisconsin-Madison, Department of Neurology, Madison, WI, USA; University of Wisconsin-Madison, Department of Medicine, Madison, WI, USA
| | - Andrzej Dekundy
- Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
| | - Andreas Gravius
- Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
| | - Wojciech Danysz
- Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
| | - Cara J Westmark
- University of Wisconsin-Madison, Department of Neurology, Madison, WI, USA.
| |
Collapse
|
27
|
Kodam A, Ourdev D, Maulik M, Hariharakrishnan J, Banerjee M, Wang Y, Kar S. A role for astrocyte-derived amyloid β peptides in the degeneration of neurons in an animal model of temporal lobe epilepsy. Brain Pathol 2018; 29:28-44. [PMID: 29665128 DOI: 10.1111/bpa.12617] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Kainic acid, an analogue of the excitatory neurotransmitter glutamate, can trigger seizures and neurotoxicity in the hippocampus and other limbic structures in a manner that mirrors the neuropathology of human temporal lobe epilepsy (TLE). However, the underlying mechanisms associated with the neurotoxicity remain unclear. Since amyloid-β (Aβ) peptides, which are critical in the development of Alzheimer's disease, can mediate toxicity by activating glutamatergic NMDA receptors, it is likely that the enhanced glutamatergic transmission that renders hippocampal neurons vulnerable to kainic acid treatment may involve Aβ peptides. Thus, we seek to establish what role Aβ plays in kainic acid-induced toxicity using in vivo and in vitro paradigms. Our results show that systemic injection of kainic acid to adult rats triggers seizures, gliosis and loss of hippocampal neurons, along with increased levels/processing of amyloid precursor protein (APP), resulting in the enhanced production of Aβ-related peptides. The changes in APP levels/processing were evident primarily in activated astrocytes, implying a role for astrocytic Aβ in kainic acid-induced toxicity. Accordingly, we showed that treating rat primary cultured astrocytes with kainic acid can lead to increased Aβ production/secretion without any compromise in cell viability. Additionally, we revealed that kainic acid reduces neuronal viability more in neuronal/astrocyte co-cultures than in pure neuronal culture, and this is attenuated by precluding Aβ production. Collectively, these results indicate that increased production/secretion of Aβ-related peptides from activated astrocytes can contribute to neurotoxicity in kainic acid-treated rats. Since kainic acid administration can lead to neuropathological changes resembling TLE, it is likely that APP/Aβ peptides derived from astrocytes may have a role in TLE pathogenesis.
Collapse
Affiliation(s)
- A Kodam
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - D Ourdev
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - M Maulik
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - J Hariharakrishnan
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - M Banerjee
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - Y Wang
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - S Kar
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| |
Collapse
|
28
|
Faundez V, De Toma I, Bardoni B, Bartesaghi R, Nizetic D, de la Torre R, Cohen Kadosh R, Herault Y, Dierssen M, Potier MC. Translating molecular advances in Down syndrome and Fragile X syndrome into therapies. Eur Neuropsychopharmacol 2018; 28:675-690. [PMID: 29887288 DOI: 10.1016/j.euroneuro.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 02/19/2018] [Accepted: 03/12/2018] [Indexed: 12/27/2022]
Abstract
Ongoing treatments for genetic developmental disorders of the central nervous system are mostly symptomatic and do not correct the genetic cause. Recent identification of common mechanisms between diseases has suggested that new therapeutic targets could be applied across intellectual disabilities with potential disease-modifying properties. The European Down syndrome and other genetic developmental disorders (DSG2D) network joined basic and clinical scientists to foster this research and carry out clinical trials. Here we discuss common mechanisms between several intellectual disabilities from genetic origin including Down's and Fragile X syndromes: i) how to model these complex diseases using neuronal cells and brain organoids derived from induced pluripotent stem cells; ii) how to integrate genomic, proteomic and interactome data to help defining common mechanisms and boundaries between diseases; iii) how to target common pathways for designing clinical trials and assessing their efficacy; iv) how to bring new neuro-therapies, such as noninvasive brain stimulations and cognitive training to clinical research. The basic and translational research efforts of the last years have utterly transformed our understanding of the molecular pathology of these diseases but much is left to be done to bring them to newborn babies and children to improve their quality of life.
Collapse
Affiliation(s)
- Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, GA, USA
| | - Ilario De Toma
- Cellular and Systems Neurobiology, Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centro de Investigación Biomédica en Red CIBERER, Spain
| | - Barbara Bardoni
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Valbonne, France
| | - Renata Bartesaghi
- University of Bologna, Department of Biomedical and Neuromotor Sciences, Bologna, Italy
| | - Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Barts and The London School of Medicine, Queen Mary University of London, United Kingdom
| | - Rafael de la Torre
- Integrated Pharmacology and Neurosciences Systems Research Group, IMIM-Hospital del Mar Medical Research Institute, Barcelona, Spain; CIBEROBN, Madrid, Spain
| | - Roi Cohen Kadosh
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Mara Dierssen
- Cellular and Systems Neurobiology, Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centro de Investigación Biomédica en Red CIBERER, Spain.
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, 47 Bd de l'Hôpital, Paris, France.
| |
Collapse
|
29
|
Prasad H, Rao R. Histone deacetylase-mediated regulation of endolysosomal pH. J Biol Chem 2018; 293:6721-6735. [PMID: 29567836 DOI: 10.1074/jbc.ra118.002025] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/19/2018] [Indexed: 11/06/2022] Open
Abstract
The pH of the endolysosomal system is tightly regulated by a balance of proton pump and leak mechanisms that are critical for storage, recycling, turnover, and signaling functions in the cell. Dysregulation of endolysosomal pH has been linked to aging, amyloidogenesis, synaptic dysfunction, and various neurodegenerative disorders, including Alzheimer's disease. Therefore, understanding the mechanisms that regulate luminal pH may be key to identifying new targets for managing these disorders. Meta-analysis of yeast microarray databases revealed that nutrient-limiting conditions inhibited the histone deacetylase (HDAC) Rpd3 and thereby up-regulated transcription of the endosomal Na+/H+ exchanger Nhx1, resulting in vacuolar alkalinization. Consistent with these findings, Rpd3 inhibition by the HDAC inhibitor and antifungal drug trichostatin A induced Nhx1 expression and vacuolar alkalinization. Bioinformatics analysis of Drosophila and mouse databases revealed that caloric control of the Nhx1 orthologs DmNHE3 and NHE6, respectively, is also mediated by HDACs. We show that NHE6 is a target of the transcription factor cAMP-response element-binding protein (CREB), a known regulator of cellular responses to low-nutrient conditions, providing a molecular mechanism for nutrient- and HDAC-dependent regulation of endosomal pH. Of note, pharmacological targeting of the CREB pathway to increase NHE6 expression helped regulate endosomal pH and correct defective clearance of amyloid Aβ in an apoE4 astrocyte model of Alzheimer's disease. These observations from yeast, fly, mouse, and cell culture models point to an evolutionarily conserved mechanism for HDAC-mediated regulation of endosomal NHE expression. Our insights offer new therapeutic strategies for modulation of endolysosomal pH in fungal infection and human disease.
Collapse
Affiliation(s)
- Hari Prasad
- From the Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Rajini Rao
- From the Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
30
|
Moazen-Zadeh E, Shirzad F, Karkhaneh-Yousefi MA, Khezri R, Mohammadi MR, Akhondzadeh S. Simvastatin as an Adjunctive Therapy to Risperidone in Treatment of Autism: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. J Child Adolesc Psychopharmacol 2018; 28:82-89. [PMID: 28719227 DOI: 10.1089/cap.2017.0055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Providing novel treatments for autism has been a subject of long-standing research. Based on etiopathological findings, we aim at assessing potential therapeutic effects of statins, here simvastatin, on autism symptoms for the first time. METHODS In this randomized, double-blind, placebo-controlled, parallel-group 10-week clinical trial, 70 drug-free children aged 4 to 12 years old with diagnosis of autistic disorder based on the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision, who had an Aberrant Behavior Checklist-Community (ABC-C) scale irritability subscale score of ≥12, were equally randomized to receive either simvastatin (20-40 mg/day) or placebo as an adjunct to risperidone (1-2 mg/day) whereas administration of both drugs was started simultaneously from baseline. Patients with comorbid psychiatric disorders, active medical conditions, severe intellectual disability, seizure disorders, history of any treatments for autism in the past 6 months, or history of current anti-inflammatory drug consumption were excluded. Primary outcome was defined as the difference in mean change of the ABC-C scale irritability subscale score from baseline to the endpoint ( www.irct.ir ; IRCT201602041556N86). RESULTS Significant differences in change of the ABC-C scale irritability (mean difference [95% confidence interval (CI)] = -3.45 [-5.37 to -1.54], p = 0.001; Cohen's d = 0.89) and hyperactivity/noncompliance (mean difference [95% CI] = -4.27 [-6.69 to -1.86], p = 0.001; Cohen's d = 0.87) subscales scores were detected between the two arms. No significant difference was detected in case of the other three subscales. CONCLUSIONS This study provides preliminary evidence for potential therapeutic effects of simvastatin in the treatment of autism that warrants further investigations.
Collapse
Affiliation(s)
- Ehsan Moazen-Zadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Fatemeh Shirzad
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | | | - Rasoul Khezri
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Mohammad-Reza Mohammadi
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences , Tehran, Iran
| |
Collapse
|
31
|
Ahn HJ, Chen ZL, Zamolodchikov D, Norris EH, Strickland S. Interactions of β-amyloid peptide with fibrinogen and coagulation factor XII may contribute to Alzheimer's disease. Curr Opin Hematol 2018; 24:427-431. [PMID: 28661939 DOI: 10.1097/moh.0000000000000368] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW To review the evidence that the Alzheimer peptide β-amyloid interacts with the blood coagulation system and influences the pathophysiology of the disease. RECENT FINDINGS β-amyloid can interact with fibrinogen and blood coagulation factor XII and trigger ischemia and inflammation. SUMMARY β-amyloid interacts with fibrinogen and factor XII. These interactions can lead to increased clotting, abnormal clot formation, persistent fibrin deposition, and generation of proinflammatory molecules. These events can damage neurons and could contribute to the cognitive decline in Alzheimer's disease patients.
Collapse
Affiliation(s)
- Hyung J Ahn
- aPatricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York City bRegeneron Pharmaceuticals, Tarrytown, New York, USA *Hyung J. Ahn, Zu-Lin Chen, and Daria Zamolodchikov contributed equally to this article
| | | | | | | | | |
Collapse
|
32
|
Arnés M, Casas-Tintó S, Malmendal A, Ferrús A. Amyloid β42 peptide is toxic to non-neural cells in Drosophila yielding a characteristic metabolite profile and the effect can be suppressed by PI3K. Biol Open 2017; 6:1664-1671. [PMID: 29141953 PMCID: PMC5703620 DOI: 10.1242/bio.029991] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The human Aβ42 peptide is associated with Alzheimer's disease through its deleterious effects in neurons. Expressing the human peptide in adult Drosophila in a tissue- and time-controlled manner, we show that Aβ42 is also toxic in non-neural cells, neurosecretory and epithelial cell types in particular. This form of toxicity includes the aberrant signaling by Wingless morphogen leading to the eventual activation of Caspase 3. Preventing Caspase 3 activation by means of p53 keeps epithelial cells from elimination but maintains the Aβ42 toxicity yielding more severe deleterious effects to the organism. Metabolic profiling by nuclear magnetic resonance (NMR) of adult flies at selected ages post Aβ42 expression onset reveals characteristic changes in metabolites as early markers of the pathological process. All morphological and most metabolic features of Aβ42 toxicity can be suppressed by the joint overexpression of PI3K. Summary: The Alzheimer's disease-related Aβ42 peptide is toxic for non-neural cells. This toxicity can be detected by specific metabolite changes and suppressed by the overexpression of the enzyme PI3K.
Collapse
Affiliation(s)
- Mercedes Arnés
- Dept. of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Avda. Doctor Arce, 37, 28002 Madrid, Spain
| | - Sergio Casas-Tintó
- Dept. of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Avda. Doctor Arce, 37, 28002 Madrid, Spain
| | - Anders Malmendal
- Biochemistry and Structural Biology, Center for Molecular Protein Science, Department of Chemistry, Lund University, P.O. Box 124, SE-22100 Lund, Sweden
| | - Alberto Ferrús
- Dept. of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Avda. Doctor Arce, 37, 28002 Madrid, Spain
| |
Collapse
|
33
|
Castagnola S, Bardoni B, Maurin T. The Search for an Effective Therapy to Treat Fragile X Syndrome: Dream or Reality? Front Synaptic Neurosci 2017; 9:15. [PMID: 29163124 PMCID: PMC5681520 DOI: 10.3389/fnsyn.2017.00015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/16/2017] [Indexed: 12/22/2022] Open
Abstract
Fragile X Syndrome (FXS) is the most common form of intellectual disability and a primary cause of autism. It originates from the lack of the Fragile X Mental Retardation Protein (FMRP), which is an RNA-binding protein encoded by the Fragile X Mental Retardation Gene 1 (FMR1) gene. Multiple roles have been attributed to this protein, ranging from RNA transport (from the nucleus to the cytoplasm, but also along neurites) to translational control of mRNAs. Over the last 20 years many studies have found a large number of FMRP mRNA targets, but it is still not clear which are those playing a critical role in the etiology of FXS. So far, no therapy for FXS has been found, making the quest for novel targets of considerable importance. Several pharmacological approaches have been attempted, but, despite some promising preclinical results, no strategy gave successful outcomes, due either to the induction of major side effects or to the lack of improvement of the phenotypes. However, these studies suggested that, in order to measure the effectiveness of a specific treatment, trials should be redesigned and new endpoints defined in FXS patients. Nevertheless, the search for new therapeutic targets for FXS is very active. In this context, the advances in animal modeling, coupled with better understanding of neurobiology and physiopathology of FXS, are of crucial importance in developing new selected treatments. Here, we discuss the pathways that were recently linked to the physiopathology of FXS (mGluR, GABAR, insulin, Insulin-like Growth Factor 1 (IGF-1), MPP-9, serotonin, oxytocin and endocannabinoid signaling) and that suggest new approaches to find an effective therapy for this disorder. Our goal with this review article is to summarize some recent relevant findings on FXS treatment strategies in order to have a clearer view of the different pathways analyzed to date emphasizing those shared with other synaptic disorders.
Collapse
Affiliation(s)
- Sara Castagnola
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Thomas Maurin
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| |
Collapse
|
34
|
Westmark CJ. Commentary: Depletion of the Fragile X Mental Retardation Protein in Embryonic Stem Cells Alters the Kinetics of Neurogenesis. Front Mol Neurosci 2017; 10:29. [PMID: 28223919 PMCID: PMC5293769 DOI: 10.3389/fnmol.2017.00029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/24/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Cara J Westmark
- Department of Neurology, University of Wisconsin Madison, WI, USA
| |
Collapse
|
35
|
Bardoni B, Capovilla M, Lalli E. Modeling Fragile X syndrome in neurogenesis: An unexpected phenotype and a novel tool for future therapies. NEUROGENESIS 2017; 4:e1270384. [PMID: 28203608 DOI: 10.1080/23262133.2016.1270384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/30/2016] [Accepted: 12/02/2016] [Indexed: 10/20/2022]
Abstract
FMRP is an RNA-binding protein involved in synaptic translation. Its absence causes a form of intellectual disability, the Fragile X syndrome (FXS). Small neuroanatomical abnormalities, present both in human and mouse FMRP-deficient brains, suggest a subtle critical role of this protein in neurogenesis. Stable depletion of FMRP has been obtained in a mouse embryonic stem cell line Fmr1 (shFmr1 ES) that does not display morphological alterations, but an abnormal expression of a subset of genes mainly involved in neuronal differentiation and maturation. Inducing the differentiation of shFmr1 ES cells into the neuronal lineage results in an accelerated generation of neural progenitors and neurons during the first steps of neurogenesis. This transient phenotype is due to an elevated level of the Amyloid Precursor Protein (APP), whose mRNA is a target of FMRP. APP is processed by the BACE-1 enzyme, producing the β-amyloid (Aβ) peptide accelerating neurogenesis by activating the expression of Ascll. Inhibition of the BACE-1 enzyme rescues the phenotype of shFmr1 ES cells. Here we discuss the importance of the shFmr1 ES line not only to understand the physiopathology of FXS but also as a tool to screen biomolecules for new FXS therapies.
Collapse
Affiliation(s)
- Barbara Bardoni
- Université Côte d'Azur, Nice, France; CNRS UMR7275, Institut of Molecular and Cellular Pharmacology, Valbonne, France; CNRS LIA « NEOGENEX », Valbonne, France
| | - Maria Capovilla
- Université Côte d'Azur, Nice, France; UMR 1355-7254 INRA/CNRS, Institut Sophia Agrobiotech, Sophia Antipolis, France
| | - Enzo Lalli
- Université Côte d'Azur, Nice, France; CNRS UMR7275, Institut of Molecular and Cellular Pharmacology, Valbonne, France; CNRS LIA « NEOGENEX », Valbonne, France
| |
Collapse
|
36
|
Westmark CJ, Chuang SC, Hays SA, Filon MJ, Ray BC, Westmark PR, Gibson JR, Huber KM, Wong RKS. APP Causes Hyperexcitability in Fragile X Mice. Front Mol Neurosci 2016; 9:147. [PMID: 28018172 PMCID: PMC5156834 DOI: 10.3389/fnmol.2016.00147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/01/2016] [Indexed: 01/06/2023] Open
Abstract
Amyloid-beta protein precursor (APP) and metabolite levels are altered in fragile X syndrome (FXS) patients and in the mouse model of the disorder, Fmr1KO mice. Normalization of APP levels in Fmr1KO mice (Fmr1KO /APPHET mice) rescues many disease phenotypes. Thus, APP is a potential biomarker as well as therapeutic target for FXS. Hyperexcitability is a key phenotype of FXS. Herein, we determine the effects of APP levels on hyperexcitability in Fmr1KO brain slices. Fmr1KO /APPHET slices exhibit complete rescue of UP states in a neocortical hyperexcitability model and reduced duration of ictal discharges in a CA3 hippocampal model. These data demonstrate that APP plays a pivotal role in maintaining an appropriate balance of excitation and inhibition (E/I) in neural circuits. A model is proposed whereby APP acts as a rheostat in a molecular circuit that modulates hyperexcitability through mGluR5 and FMRP. Both over- and under-expression of APP in the context of the Fmr1KO increases seizure propensity suggesting that an APP rheostat maintains appropriate E/I levels but is overloaded by mGluR5-mediated excitation in the absence of FMRP. These findings are discussed in relation to novel treatment approaches to restore APP homeostasis in FXS.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin-Madison, MadisonMadison, WI, USA
| | - Shih-Chieh Chuang
- Department of Physiology and Pharmacology, State University of New York Downstate Medical CenterBrooklyn, NY, USA
| | - Seth A. Hays
- Department of Neuroscience, University of Texas Southwestern Medical CenterDallas, TX, USA
| | - Mikolaj J. Filon
- Department of Neurology, University of Wisconsin-Madison, MadisonMadison, WI, USA
| | - Brian C. Ray
- Department of Neurology, University of Wisconsin-Madison, MadisonMadison, WI, USA
| | - Pamela R. Westmark
- Department of Medicine, University of Wisconsin-Madison, MadisonMadison, WI, USA
| | - Jay R. Gibson
- Department of Neuroscience, University of Texas Southwestern Medical CenterDallas, TX, USA
| | - Kimberly M. Huber
- Department of Neuroscience, University of Texas Southwestern Medical CenterDallas, TX, USA
| | - Robert K. S. Wong
- Department of Physiology and Pharmacology, State University of New York Downstate Medical CenterBrooklyn, NY, USA
| |
Collapse
|