1
|
Treccarichi S, Vinci M, Musumeci A, Rando RG, Papa C, Saccone S, Federico C, Failla P, Ruggieri M, Calì F, Polizzi A, Praticò A. Investigating the Role of the Zinc Finger Protein ZC2HC1C on Autism Spectrum Disorder Susceptibility. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:574. [PMID: 40282865 PMCID: PMC12029121 DOI: 10.3390/medicina61040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Zinc finger proteins are important transcription factors that regulate gene expression and play a critical role in neurodevelopment including autism spectrum disorders (ASDs). They are involved in a variety of cellular processes, including cell proliferation, differentiation, and apoptosis. Materials and Methods: Whole-exome sequencing (WES) analysis on a patient diagnosed with ASD. Results: Sequencing identified a homozygous insertion causing a stop codon, resulting in the removal of several functional domains including the zinc finger C2HC/C3H type of the ZC2HC1C protein. To date, no MIM entry has been assigned to the detected gene. In silico predictions described the variant as likely pathogenic, indicating an autosomal recessive inheritance pattern. In this study, we hypothesize that this homozygous mutation disrupts protein function and may represent a susceptibility gene for autism. The parents and the patient's sister were healthy and carry the variant in the heterozygous condition. This gene is expressed in brain tissues showing high expression in both the choroid plexus (ChP) and midbrain, whose dysfunctions, as reported, may lead to ASD. Moreover, predictive pathway analyses indicated the probable involvement of this gene in primary cilia development. This process has been frequently linked to neurodevelopmental impairments, such as autism, as documented in previous studies. Conclusions: Further analyses are needed via in vitro functional assays or by ZC2HC1C gene knockout to validate its functional role.
Collapse
Affiliation(s)
- Simone Treccarichi
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Mirella Vinci
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Antonino Musumeci
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Rosanna Galati Rando
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Carla Papa
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Salvatore Saccone
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy; (S.S.); (C.F.)
| | - Concetta Federico
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy; (S.S.); (C.F.)
| | - Pinella Failla
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Martino Ruggieri
- Unit of Pediatric Clinic, Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia 89, 95123 Catania, Italy;
| | - Francesco Calì
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (S.T.); (M.V.); (A.M.); (R.G.R.); (C.P.); (P.F.)
| | - Agata Polizzi
- Department of Formative Process, University of Catania, Via Teatro Greco 84, 95124 Catania, Italy;
| | - Andrea Praticò
- Deparment of Medicine and Surgery, University Kore of Enna, Cittadella Universitaria, 94100 Enna, Italy;
| |
Collapse
|
2
|
Zhong R, Chernick D, Hottman D, Tan Y, Kim M, Narayanan M, Li L. The HDL-Mimetic Peptide 4F Mitigates Vascular and Cortical Amyloid Pathology and Associated Neuroinflammation in a Transgenic Mouse Model of Cerebral Amyloid Angiopathy and Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04859-9. [PMID: 40120042 DOI: 10.1007/s12035-025-04859-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Despite recent advances, more effective and safer treatment options for AD are needed. Cerebral amyloid angiopathy (CAA) is one of the key pathological hallmarks of AD characterized by amyloid-β (Aβ) deposition in the cerebral vasculature and is associated with intracerebral hemorrhage, cerebrovascular dysfunction, and cognitive impairment. CAA is also considered to underlie the main adverse effect of recently FDA-approved anti-Aβ immunotherapies, namely the amyloid-related imaging abnormalities (ARIA). Substantial evidence has shown that elevated levels of high-density lipoprotein (HDL) and its main protein component, APOA-I, are associated with reduced CAA and superior cognitive function. 4F is an APOA-I/HDL-mimetic peptide and its clinical safety and activity have been demonstrated in human trials for cardiovascular diseases. The present study investigates whether treatment with 4F modulates CAA and associated cognitive deficits and neuropathologies in the well-established Tg-SwDI mouse model of CAA/AD. Age/sex-matched Tg-SwDI mice received daily treatments of 4F or vehicle (PBS), respectively, by intraperitoneal injections for 12 weeks. The results showed that 4F treatment reduced overall Aβ plaque deposition and CAA, and attenuated CAA-associated microgliosis, without significantly affecting total levels of Aβ, astrocytosis, and behavioral function. Unbiased transcriptomic analysis revealed a heightened inflammatory state in the brain of SwDI mice and that 4F treatment reversed the overactivation of vascular cells, in particular vascular smooth muscle cells, relieving cerebrovascular inflammation in CAA/AD mice. Our study provides experimental evidence for the therapeutic potential of 4F to mitigate CAA and associated pathologies in AD.
Collapse
Affiliation(s)
- Rui Zhong
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Dustin Chernick
- Graduate Program in Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - David Hottman
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yejun Tan
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Minwoo Kim
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Manojkumar Narayanan
- Graduate Program in Comparative and Molecular Biosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Graduate Program in Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
3
|
Zheng J, Wang H, Wu W, Wang L, Qin M, Zhu L, Liu Z, Chen Y, Yu Y. Role of FPR2 antagonism in alleviating social isolation-induced depression and protecting blood-brain barrier integrity. J Neuroinflammation 2025; 22:79. [PMID: 40083006 PMCID: PMC11907847 DOI: 10.1186/s12974-025-03408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Social isolation (SI) is a prevalent issue in modern society, particularly exacerbated during the COVID-19 pandemic, and it is a significant contributor to depressive disorders. Inflammation-related markers are upregulated in patients with major depressive disorder (MDD) unresponsive to first-line selective serotonin reuptake inhibitor (SSRI) antidepressants. This study investigates the role of formyl peptide receptor 2 (FPR2), a G-protein coupled receptor expressed in central and peripheral immune cells, in SI-induced depression. We developed a mouse model of SI by housing mice individually for three weeks. SI mice exhibited increased capillary-associated microglia (CAMs) with upregulated FPR2 expression in the prefrontal cortex (PFC) and hippocampus compared to group-housed controls. Notably, subcutaneous administration of the FPR2 antagonist WRW4 alleviated depressive and anxiety-like behaviors in SI mice, reducing microglial activation and neuronal damage. WRW4 treatment decreased CAM numbers and their FPR2 expression. RNA sequencing revealed that SI primarily induced changes in genes associated with blood-brain barrier (BBB) function, followed by alterations in genes related to hormone activity, immune activation, and neuronal function. Transcriptomic changes in brain endothelial cells from SI mice resembled those observed in animal models of several neurological disorders and in MDD patients. WRW4 treatment partially reversed these transcriptomic alterations and restored compromised BBB integrity. Additionally, intracerebroventricular (ICV) injection of WRW4 also alleviated depressive and anxiety-like behaviors in SI mice. Finally, our analysis of public transcriptome databases indicates FPR2 upregulation in the orbital ventral PFC of MDD patients and peripheral blood mononuclear cells of those in severe depressive episodes. These findings suggest that the pharmacological targeting of FPR2 may rescue SI-induced pathology in mice by protecting BBB integrity.
Collapse
Affiliation(s)
- Jiayi Zheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hanqi Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanning Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Linlin Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Meizhen Qin
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, China
| | - Lingfeng Zhu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhen Liu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijun Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
4
|
Khan A, Minbay M, Attia Z, Ay AA, Ingram KK. Sex- and Substance-Specific Associations of Circadian-Related Genes with Addiction in the UK Biobank Cohort Implicate Neuroplasticity Pathways. Brain Sci 2024; 14:1282. [PMID: 39766481 PMCID: PMC11674644 DOI: 10.3390/brainsci14121282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES The circadian clockwork is implicated in the etiology of addiction, with circadian rhythm disruptions bidirectionally linked to substance abuse, but the molecular mechanisms that underlie this connection are not well known. METHODS Here, we use machine learning to reveal sex- and substance-specific associations with addiction in variants from 51 circadian-related genes (156,702 SNPs) in 98,800 participants from a UK Biobank cohort. We further analyze SNP associations in a subset of the cohort for substance-specific addictions (alcohol, illicit drugs (narcotics), and prescription drugs (opioids)). RESULTS We find robust (OR > 10) and novel sex-specific and substance-specific associations with variants in synaptic transcription factors (ZBTB20, CHRNB3) and hormone receptors (RORA), particularly in individuals addicted to narcotics and opioids. Circadian-related gene variants associated with male and female addiction were non-overlapping; variants in males primarily involve dopaminergic pathways, while variants in females factor in metabolic and inflammation pathways, with a novel gene association of female addiction with DELEC1, a gene of unknown function. CONCLUSIONS Our findings underscore the complexity of genetic pathways associated with addiction, involving core clock genes and circadian-regulated pathways, and reveal novel circadian-related gene associations that will aid the development of targeted, sex-specific therapeutic interventions for substance abuse.
Collapse
Affiliation(s)
- Ayub Khan
- Department of Biology, Colgate University, Hamilton, NY 13346, USA; (A.K.); (A.A.A.)
- Department of Computer Science, Colgate University, Hamilton, NY 13346, USA; (M.M.); (Z.A.)
| | - Mete Minbay
- Department of Computer Science, Colgate University, Hamilton, NY 13346, USA; (M.M.); (Z.A.)
| | - Ziad Attia
- Department of Computer Science, Colgate University, Hamilton, NY 13346, USA; (M.M.); (Z.A.)
- Department of Mathematics, Colgate University, Hamilton, NY 13346, USA
| | - Ahmet Ali Ay
- Department of Biology, Colgate University, Hamilton, NY 13346, USA; (A.K.); (A.A.A.)
- Department of Mathematics, Colgate University, Hamilton, NY 13346, USA
| | - Krista K. Ingram
- Department of Biology, Colgate University, Hamilton, NY 13346, USA; (A.K.); (A.A.A.)
| |
Collapse
|
5
|
Moon YM, Park SE, Smith-Hicks C, Hauptman A. Resolution of severe neurobehavioral difficulties in an individual with Primrose syndrome with sertraline. Am J Med Genet A 2024; 194:e63610. [PMID: 38517161 DOI: 10.1002/ajmg.a.63610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/23/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Primrose syndrome (PS) is a rare genetic disease characterized by developmental delay, intellectual disability, sensorineural hearing loss, and dysmorphic features. PS is caused by de novo pathogenic variants in the ZBTB20 gene, which encodes a transcription factor modulating neurogenesis. We describe resolution with sertraline of neurobehavioral difficulties in a 17-year-old Hispanic male with PS with de novo heterozygous c.1916G > A (p.C639Y) variant of ZBTB20. Neurobehavioral difficulties included aggression towards self and others, irritability, tearfulness, and mood liability that did not respond to behavioral interventions or aripiprazole. Treatment with sertraline, a medication indicated for psychiatric disorders including anxiety and depression, led to the resolution of neurobehavioral difficulties after 2 weeks of initiation of medication. The treatment course suggests that selective serotonin reuptake inhibitors, such as sertraline, may be a useful tool for neurobehavioral difficulties in PS over antipsychotics that are accompanied by complex side effect profiles, and suggest that anxiety is the primary cause of the neurobehavioral difficulties in this patient.
Collapse
Affiliation(s)
- Young Min Moon
- Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sa Eun Park
- Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Psychiatry, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Constance Smith-Hicks
- Department of Neurology, School of Medicine, Johns Hopkins Univerisity, Baltimore, Maryland, USA
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Aaron Hauptman
- Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Psychiatry, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Cummins M, Watson C, Edwards RJ, Mattick JS. The Evolution of Ultraconserved Elements in Vertebrates. Mol Biol Evol 2024; 41:msae146. [PMID: 39058500 PMCID: PMC11276968 DOI: 10.1093/molbev/msae146] [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: 03/26/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Ultraconserved elements were discovered two decades ago, arbitrarily defined as sequences that are identical over a length ≥ 200 bp in the human, mouse, and rat genomes. The definition was subsequently extended to sequences ≥ 100 bp identical in at least three of five mammalian genomes (including dog and cow), and shown to have undergone rapid expansion from ancestors in fish and strong negative selection in birds and mammals. Since then, many more genomes have become available, allowing better definition and more thorough examination of ultraconserved element distribution and evolutionary history. We developed a fast and flexible analytical pipeline for identifying ultraconserved elements in multiple genomes, dedUCE, which allows manipulation of minimum length, sequence identity, and number of species with a detectable ultraconserved element according to specified parameters. We suggest an updated definition of ultraconserved elements as sequences ≥ 100 bp and ≥97% sequence identity in ≥50% of placental mammal orders (12,813 ultraconserved elements). By mapping ultraconserved elements to ∼200 species, we find that placental ultraconserved elements appeared early in vertebrate evolution, well before land colonization, suggesting that the evolutionary pressures driving ultraconserved element selection were present in aquatic environments in the Cambrian-Devonian periods. Most (>90%) ultraconserved elements likely appeared after the divergence of gnathostomes from jawless predecessors, were largely established in sequence identity by early Sarcopterygii evolution-before the divergence of lobe-finned fishes from tetrapods-and became near fixed in the amniotes. Ultraconserved elements are mainly located in the introns of protein-coding and noncoding genes involved in neurological and skeletomuscular development, enriched in regulatory elements, and dynamically expressed throughout embryonic development.
Collapse
Affiliation(s)
- Mitchell Cummins
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Cadel Watson
- School of Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Richard J Edwards
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - John S Mattick
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
7
|
Chato-Astrain I, Pronot M, Coppola T, Martin S. Molecular Organization and Regulation of the Mammalian Synapse by the Post-Translational Modification SUMOylation. Cells 2024; 13:420. [PMID: 38474384 PMCID: PMC10930594 DOI: 10.3390/cells13050420] [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: 02/02/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Neurotransmission occurs within highly specialized compartments forming the active synapse where the complex organization and dynamics of the interactions are tightly orchestrated both in time and space. Post-translational modifications (PTMs) are central to these spatiotemporal regulations to ensure an efficient synaptic transmission. SUMOylation is a dynamic PTM that modulates the interactions between proteins and consequently regulates the conformation, the distribution and the trafficking of the SUMO-target proteins. SUMOylation plays a crucial role in synapse formation and stabilization, as well as in the regulation of synaptic transmission and plasticity. In this review, we summarize the molecular consequences of this protein modification in the structural organization and function of the mammalian synapse. We also outline novel activity-dependent regulation and consequences of the SUMO process and explore how this protein modification can functionally participate in the compartmentalization of both pre- and post-synaptic sites.
Collapse
Affiliation(s)
- Isabel Chato-Astrain
- Université Côte d’Azur, CNRS, Inserm, IPMC, Sophia Antipolis, F-06560 Valbonne, France; (I.C.-A.); (T.C.)
| | - Marie Pronot
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK;
| | - Thierry Coppola
- Université Côte d’Azur, CNRS, Inserm, IPMC, Sophia Antipolis, F-06560 Valbonne, France; (I.C.-A.); (T.C.)
| | - Stéphane Martin
- Université Côte d’Azur, CNRS, Inserm, IPMC, Sophia Antipolis, F-06560 Valbonne, France; (I.C.-A.); (T.C.)
| |
Collapse
|
8
|
Ward SK, Wadley A, Tsai CHA, Benke PJ, Emrick L, Fisher K, Houck KM, Dai H, Guillen Sacoto MJ, Craigen W, Glaser K, Murdock DR, Rohena L, Diderich KEM, Bruggenwirth HT, Lee B, Bacino C, Burrage LC, Rosenfeld JA. De novo missense variants in ZBTB47 are associated with developmental delays, hypotonia, seizures, gait abnormalities, and variable movement abnormalities. Am J Med Genet A 2024; 194:17-30. [PMID: 37743782 PMCID: PMC11221546 DOI: 10.1002/ajmg.a.63399] [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: 03/12/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023]
Abstract
The collection of known genetic etiologies of neurodevelopmental disorders continues to increase, including several syndromes associated with defects in zinc finger protein transcription factors (ZNFs) that vary in clinical severity from mild learning disabilities and developmental delay to refractory seizures and severe autism spectrum disorder. Here we describe a new neurodevelopmental disorder associated with variants in ZBTB47 (also known as ZNF651), which encodes zinc finger and BTB domain-containing protein 47. Exome sequencing (ES) was performed for five unrelated patients with neurodevelopmental disorders. All five patients are heterozygous for a de novo missense variant in ZBTB47, with p.(Glu680Gly) (c.2039A>G) detected in one patient and p.(Glu477Lys) (c.1429G>A) identified in the other four patients. Both variants impact conserved amino acid residues. Bioinformatic analysis of each variant is consistent with pathogenicity. We present five unrelated patients with de novo missense variants in ZBTB47 and a phenotype characterized by developmental delay with intellectual disability, seizures, hypotonia, gait abnormalities, and variable movement abnormalities. We propose that these variants in ZBTB47 are the basis of a new neurodevelopmental disorder.
Collapse
Affiliation(s)
- Scott K Ward
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
- Department of Pediatrics, Division of Medical Genetics and Genomic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alexandrea Wadley
- Department of Pediatrics, Section of Genetics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Chun-Hui Anne Tsai
- Department of Pediatrics, Section of Genetics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Paul J Benke
- Joe DiMaggio Children's Hospital, Hollywood, Florida, USA
| | - Lisa Emrick
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine (BCM), Houston, Texas, USA
| | - Kristen Fisher
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine (BCM), Houston, Texas, USA
| | - Kimberly M Houck
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine (BCM), Houston, Texas, USA
| | - Hongzheng Dai
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
| | | | - William Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
| | - Kimberly Glaser
- Joe DiMaggio Children's Hospital, Hollywood, Florida, USA
- Invitae, San Francisco, California, USA
| | - David R Murdock
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Luis Rohena
- Department of Pediatrics, Division of Medical Genetics, San Antonio Military Medical Center, San Antonio, Texas, USA
- Department of Pediatrics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Karin E M Diderich
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Hennie T Bruggenwirth
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
| | - Carlos Bacino
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, USA
| |
Collapse
|
9
|
Kjær C, Palasca O, Barzaghi G, Bak LK, Durhuus RKJ, Jakobsen E, Pedersen L, Bartels ED, Woldbye DPD, Pinborg LH, Jensen LJ. Differential Expression of the β3 Subunit of Voltage-Gated Ca 2+ Channel in Mesial Temporal Lobe Epilepsy. Mol Neurobiol 2023; 60:5755-5769. [PMID: 37341859 PMCID: PMC10471638 DOI: 10.1007/s12035-023-03426-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
The purpose of this study was to identify and validate new putative lead drug targets in drug-resistant mesial temporal lobe epilepsy (mTLE) starting from differentially expressed genes (DEGs) previously identified in mTLE in humans by transcriptome analysis. We identified consensus DEGs among two independent mTLE transcriptome datasets and assigned them status as "lead target" if they (1) were involved in neuronal excitability, (2) were new in mTLE, and (3) were druggable. For this, we created a consensus DEG network in STRING and annotated it with information from the DISEASES database and the Target Central Resource Database (TCRD). Next, we attempted to validate lead targets using qPCR, immunohistochemistry, and Western blot on hippocampal and temporal lobe neocortical tissue from mTLE patients and non-epilepsy controls, respectively. Here we created a robust, unbiased list of 113 consensus DEGs starting from two lists of 3040 and 5523 mTLE significant DEGs, respectively, and identified five lead targets. Next, we showed that CACNB3, a voltage-gated Ca2+ channel subunit, was significantly regulated in mTLE at both mRNA and protein level. Considering the key role of Ca2+ currents in regulating neuronal excitability, this suggested a role for CACNB3 in seizure generation. This is the first time changes in CACNB3 expression have been associated with drug-resistant epilepsy in humans, and since efficient therapeutic strategies for the treatment of drug-resistant mTLE are lacking, our finding might represent a step toward designing such new treatment strategies.
Collapse
Affiliation(s)
- Christina Kjær
- Biomedical Laboratory Science, Department of Technology, Faculty of Health and Technology, University College Copenhagen, Sigurdsgade 26, 1St, 2200 Copenhagen, Denmark
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Oana Palasca
- Disease Systems Biology Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Guido Barzaghi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Lasse K. Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Dept. of Clinical Biochemistry, 2600 RigshospitaletCopenhagen, Denmark
| | - Rúna K. J. Durhuus
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Specific Pharma A/S, Borgmester Christiansens Gade 40, 2450 Copenhagen, SV Denmark
| | - Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Takeda Pharma A/S, Delta Park 45, 2665 Vallensbaek Strand, Denmark
| | - Louise Pedersen
- Biomedical Laboratory Science, Department of Technology, Faculty of Health and Technology, University College Copenhagen, Sigurdsgade 26, 1St, 2200 Copenhagen, Denmark
- Dept. of Clinical Biochemistry, 2600 RigshospitaletCopenhagen, Denmark
| | - Emil D. Bartels
- Dept. of Clinical Biochemistry, 2600 RigshospitaletCopenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David P. D. Woldbye
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lars H. Pinborg
- Epilepsy Clinic & Neurobiology Research Unit, Copenhagen University Hospital, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Lars Juhl Jensen
- Disease Systems Biology Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Stoyanov D, Stoyanov GS, Ivanov MN, Spasov RH, Tonchev AB. Transcription Factor Zbtb20 as a Regulator of Malignancy and Its Practical Applications. Int J Mol Sci 2023; 24:13763. [PMID: 37762065 PMCID: PMC10530547 DOI: 10.3390/ijms241813763] [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: 08/03/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Zbtb20 (zinc finger and BTB domain-containing protein 20) is a transcription factor with a zinc finger DNA binding domain and a BTB domain responsible for protein-protein interaction. Recently, this TF has received attention because new data showed its pivotal involvement in normal neural development and its regulatory effects on proliferation and differentiation in different tissues. Zbtb20 was shown to increase proliferation and migration and confer resistance to apoptosis in the contexts of many malignant tumors like hepatocellular carcinoma, non-small-cell lung carcinoma, gastric adenocarcinoma, glioblastoma multiforme, breast cancer, and acute myeloid leukemia. The involvement of Zbtb20 in tumor biology is best studied in hepatocellular carcinoma, where it is a promising candidate as an immunohistochemical tumor marker or may be used in patient screening. Here we review the current data connecting Zbtb20 with malignant tumors.
Collapse
Affiliation(s)
- Dimo Stoyanov
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
| | - George S. Stoyanov
- Department of Clinical Pathology, Complex Oncology Center, 9700 Shumen, Bulgaria
| | - Martin N. Ivanov
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| | - Radoslav H. Spasov
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
| | - Anton B. Tonchev
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| |
Collapse
|
11
|
Mohan K, Gasparoni G, Salhab A, Orlich MM, Geffers R, Hoffmann S, Adams RH, Walter J, Nordheim A. Age-Associated Changes in Endothelial Transcriptome and Epigenetic Landscapes Correlate With Elevated Risk of Cerebral Microbleeds. J Am Heart Assoc 2023; 12:e031044. [PMID: 37609982 PMCID: PMC10547332 DOI: 10.1161/jaha.123.031044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Background Stroke is a leading global cause of human death and disability, with advanced aging associated with elevated incidences of stroke. Despite high mortality and morbidity of stroke, the mechanisms leading to blood-brain barrier dysfunction and development of stroke with age are poorly understood. In the vasculature of brain, endothelial cells (ECs) constitute the core component of the blood-brain barrier and provide a physical barrier composed of tight junctions, adherens junctions, and basement membrane. Methods and Results We show, in mice, the incidents of intracerebral bleeding increases with age. After isolating an enriched population of cerebral ECs from murine brains at 2, 6, 12, 18, and 24 months, we studied age-associated changes in gene expression. The study reveals age-dependent dysregulation of 1388 genes, including many involved in the maintenance of the blood-brain barrier and vascular integrity. We also investigated age-dependent changes on the levels of CpG methylation and accessible chromatin in cerebral ECs. Our study reveals correlations between age-dependent changes in chromatin structure and gene expression, whereas the dynamics of DNA methylation changes are different. Conclusions We find significant age-dependent downregulation of the Aplnr gene along with age-dependent reduction in chromatin accessibility of promoter region of the Aplnr gene in cerebral ECs. Aplnr is associated with positive regulation of vasodilation and is implicated in vascular health. Altogether, our data suggest a potential role of the apelinergic axis involving the ligand apelin and its receptor to be critical in maintenance of the blood-brain barrier and vascular integrity.
Collapse
Affiliation(s)
- Kshitij Mohan
- Interfaculty Institute of Cell BiologyUniversity of TübingenTübingenGermany
- International Max Planck Research School “From Molecules to Organisms”TübingenGermany
| | | | | | - Michael M. Orlich
- Interfaculty Institute of Cell BiologyUniversity of TübingenTübingenGermany
- International Max Planck Research School “From Molecules to Organisms”TübingenGermany
| | - Robert Geffers
- Genome AnalyticsHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Steve Hoffmann
- Leibniz Institute on AgingFritz Lipmann InstituteJenaGermany
| | - Ralf H. Adams
- Department of Tissue MorphogenesisMax Planck Institute for Molecular BiomedicineMünsterGermany
- Faculty of MedicineUniversity of MünsterMünsterGermany
| | - Jörn Walter
- Department of GeneticsUniversity of SaarlandSaarbrückenGermany
| | - Alfred Nordheim
- Interfaculty Institute of Cell BiologyUniversity of TübingenTübingenGermany
- Leibniz Institute on AgingFritz Lipmann InstituteJenaGermany
- International Max Planck Research School “From Molecules to Organisms”TübingenGermany
| |
Collapse
|
12
|
Niu M, Cao W, Wang Y, Zhu Q, Luo J, Wang B, Zheng H, Weitz DA, Zong C. Droplet-based transcriptome profiling of individual synapses. Nat Biotechnol 2023; 41:1332-1344. [PMID: 36646931 DOI: 10.1038/s41587-022-01635-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/06/2022] [Indexed: 01/17/2023]
Abstract
Synapses are crucial structures that mediate signal transmission between neurons in complex neural circuits and display considerable morphological and electrophysiological heterogeneity. So far we still lack a high-throughput method to profile the molecular heterogeneity among individual synapses. In the present study, we develop a droplet-based single-cell (sc) total-RNA-sequencing platform, called Multiple-Annealing-and-Tailing-based Quantitative scRNA-seq in Droplets, for transcriptome profiling of individual neurites, primarily composed of synaptosomes. In the synaptosome transcriptome, or 'synaptome', profiling of both mouse and human brain samples, we detect subclusters among synaptosomes that are associated with neuronal subtypes and characterize the landscape of transcript splicing that occurs within synapses. We extend synaptome profiling to synaptopathy in an Alzheimer's disease (AD) mouse model and discover AD-associated synaptic gene expression changes that cannot be detected by single-nucleus transcriptome profiling. Overall, our results show that this platform provides a high-throughput, single-synaptosome transcriptome profiling tool that will facilitate future discoveries in neuroscience.
Collapse
Affiliation(s)
- Muchun Niu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Wenjian Cao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, China
| | - Yongcheng Wang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Wyss Institute of Bioinspired Engineering, Harvard University, Cambridge, MA, USA
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Qiangyuan Zhu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, China
| | - Jiayi Luo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Baiping Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Hui Zheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - David A Weitz
- Wyss Institute of Bioinspired Engineering, Harvard University, Cambridge, MA, USA.
- Department of Physics and School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Chenghang Zong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Kim S, Oh H, Choi SH, Yoo YE, Noh YW, Cho Y, Im GH, Lee C, Oh Y, Yang E, Kim G, Chung WS, Kim H, Kang H, Bae Y, Kim SG, Kim E. Postnatal age-differential ASD-like transcriptomic, synaptic, and behavioral deficits in Myt1l-mutant mice. Cell Rep 2022; 40:111398. [PMID: 36130507 DOI: 10.1016/j.celrep.2022.111398] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/28/2022] [Accepted: 08/31/2022] [Indexed: 12/29/2022] Open
Abstract
Myelin transcription factor 1 like (Myt1l), a zinc-finger transcription factor, promotes neuronal differentiation and is implicated in autism spectrum disorder (ASD) and intellectual disability. However, it remains unclear whether Myt1l promotes neuronal differentiation in vivo and its deficiency in mice leads to disease-related phenotypes. Here, we report that Myt1l-heterozygous mutant (Myt1l-HT) mice display postnatal age-differential ASD-related phenotypes: newborn Myt1l-HT mice, with strong Myt1l expression, show ASD-like transcriptomic changes involving decreased synaptic gene expression and prefrontal excitatory synaptic transmission and altered righting reflex. Juvenile Myt1l-HT mice, with markedly decreased Myt1l expression, display reverse ASD-like transcriptomes, increased prefrontal excitatory transmission, and largely normal behaviors. Adult Myt1l-HT mice show ASD-like transcriptomes involving astrocytic and microglial gene upregulation, increased prefrontal inhibitory transmission, and behavioral deficits. Therefore, Myt1l haploinsufficiency leads to ASD-related phenotypes in newborn mice, which are temporarily normalized in juveniles but re-appear in adults, pointing to continuing phenotypic changes long after a marked decrease of Myt1l expression in juveniles.
Collapse
Affiliation(s)
- Seongbin Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hyoseon Oh
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Sang Han Choi
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Ye-Eun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Young Woo Noh
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Yisul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Chanhee Lee
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Yusang Oh
- Department of Bio and Brain Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Esther Yang
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Gyuri Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hyun Kim
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon 34141, Korea
| | - Yongchul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea.
| |
Collapse
|
14
|
Differential Methylation Profile in Fragile X Syndrome-Prone Offspring Mice after in Utero Exposure to Lactobacillus Reuteri. Genes (Basel) 2022; 13:genes13081300. [PMID: 35893036 PMCID: PMC9331364 DOI: 10.3390/genes13081300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 01/13/2023] Open
Abstract
Environmental factors such as diet, gut microbiota, and infections have proven to have a significant role in epigenetic modifications. It is known that epigenetic modifications may cause behavioral and neuronal changes observed in neurodevelopmental disabilities, including fragile X syndrome (FXS) and autism (ASD). Probiotics are live microorganisms that provide health benefits when consumed, and in some cases are shown to decrease the chance of developing neurological disorders. Here, we examined the epigenetic outcomes in offspring mice after feeding of a probiotic organism, Lactobacillus reuteri (L. reuteri), to pregnant mother animals. In this study, we tested a cohort of Western diet-fed descendant mice exhibiting a high frequency of behavioral features and lower FMRP protein expression similar to what is observed in FXS in humans (described in a companion manuscript in this same GENES special topic issue). By investigating 17,735 CpG sites spanning the whole mouse genome, we characterized the epigenetic profile in two cohorts of mice descended from mothers treated and non-treated with L. reuteri to determine the effect of prenatal probiotic exposure on the prevention of FXS-like symptoms. We found several genes involved in different neurological pathways being differentially methylated (p ≤ 0.05) between the cohorts. Among the key functions, synaptogenesis, neurogenesis, synaptic modulation, synaptic transmission, reelin signaling pathway, promotion of specification and maturation of neurons, and long-term potentiation were observed. The results of this study are relevant as they could lead to a better understanding of the pathways involved in these disorders, to novel therapeutics approaches, and to the identification of potential biomarkers for early detection of these conditions.
Collapse
|
15
|
Wang A, Wang J, Tian K, Huo D, Ye H, Li S, Zhao C, Zhang B, Zheng Y, Xu L, Hua X, Wang K, Wu QF, Wu X, Zeng T, Liu Y, Zhou Y. An epigenetic circuit controls neurogenic programs during neocortex development. Development 2021; 148:273471. [PMID: 35020876 DOI: 10.1242/dev.199772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022]
Abstract
The production and expansion of intermediate progenitors (IPs) are essential for neocortical neurogenesis during development and over evolution. Here, we have characterized an epigenetic circuit that precisely controls neurogenic programs, particularly properties of IPs, during neocortical development. The circuit comprises a long non-coding RNA (LncBAR) and the BAF (SWI/SNF) chromatin-remodeling complex, which transcriptionally maintains the expression of Zbtb20. LncBAR knockout neocortex contains more deep-layer but fewer upper-layer projection neurons. Intriguingly, loss of LncBAR promotes IP production, but paradoxically prolongs the duration of the cell cycle of IPs during mid-later neocortical neurogenesis. Moreover, in LncBAR knockout mice, depletion of the neural progenitor pool at embryonic stage results in fewer adult neural progenitor cells in the subventricular zone of lateral ventricles, leading to a failure in adult neurogenesis to replenish the olfactory bulb. LncBAR binds to BRG1, the core enzymatic component of the BAF chromatin-remodeling complex. LncBAR depletion enhances association of BRG1 with the genomic locus of, and suppresses the expression of, Zbtb20, a transcription factor gene known to regulate both embryonic and adult neurogenesis. ZBTB20 overexpression in LncBAR-knockout neural precursors reverses compromised cell cycle progressions of IPs.
Collapse
Affiliation(s)
- Andi Wang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Junbao Wang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Kuan Tian
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Dawei Huo
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China200072
| | - Hanzhe Ye
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Si Li
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China 300070
| | - Chen Zhao
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Bo Zhang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Yue Zheng
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Lichao Xu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Xiaojiao Hua
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Kun Wang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China 100101
| | - Xudong Wu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China 300070
| | - Tao Zeng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China200072
| | - Ying Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| | - Yan Zhou
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute at School of Medicine, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan, China430071
| |
Collapse
|
16
|
Medeiros de Araújo JA, Barão S, Mateos-White I, Espinosa A, Costa MR, Gil-Sanz C, Müller U. ZBTB20 is crucial for the specification of a subset of callosal projection neurons and astrocytes in the mammalian neocortex. Development 2021; 148:271200. [PMID: 34351428 DOI: 10.1242/dev.196642] [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: 09/02/2020] [Accepted: 07/17/2021] [Indexed: 12/25/2022]
Abstract
Neocortical progenitor cells generate subtypes of excitatory projection neurons in sequential order followed by the generation of astrocytes. The transcription factor zinc finger and BTB domain-containing protein 20 (ZBTB20) has been implicated in regulation of cell specification during neocortical development. Here, we show that ZBTB20 instructs the generation of a subset of callosal projections neurons in cortical layers II/III in mouse. Conditional deletion of Zbtb20 in cortical progenitors, and to a lesser degree in differentiating neurons, leads to an increase in the number of layer IV neurons at the expense of layer II/III neurons. Astrogliogenesis is also affected in the mutants with an increase in the number of a specific subset of astrocytes expressing GFAP. Astrogliogenesis is more severely disrupted by a ZBTB20 protein containing dominant mutations linked to Primrose syndrome, suggesting that ZBTB20 acts in concert with other ZBTB proteins that were also affected by the dominant-negative protein to instruct astrogliogenesis. Overall, our data suggest that ZBTB20 acts both in progenitors and in postmitotic cells to regulate cell fate specification in the mammalian neocortex.
Collapse
Affiliation(s)
- Jéssica Alves Medeiros de Araújo
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59056-450, Brazil
| | - Soraia Barão
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Isabel Mateos-White
- BIOTECMED Institute, Universidad de Valencia, Burjassot, Valencia 46100, Spain
| | - Ana Espinosa
- AntalGenics, Quorum Building III, Scientific Park - UMH. Avda. de la Universidad, s/n. 03202 Elche (Alicante), Spain
| | - Marcos Romualdo Costa
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59056-450, Brazil.,Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, U1167-Excellence Laboratory LabEx DISTALZ, Lille Cedex 59019, France
| | - Cristina Gil-Sanz
- BIOTECMED Institute, Universidad de Valencia, Burjassot, Valencia 46100, Spain
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Trakadis Y, Accogli A, Qi B, Bloom D, Joober R, Levy E, Tabbane K. Next-generation gene panel testing in adolescents and adults in a medical neuropsychiatric genetics clinic. Neurogenetics 2021; 22:313-322. [PMID: 34363551 DOI: 10.1007/s10048-021-00664-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/21/2021] [Indexed: 01/04/2023]
Abstract
Intellectual disability (ID) encompasses a clinically and genetically heterogeneous group of neurodevelopmental disorders that may present with psychiatric illness in up to 40% of cases. Despite the evidence for clinical utility of genetic panels in pediatrics, there are no published studies in adolescents/adults with ID or autism spectrum disorder (ASD). This study was approved by our institutional research ethics board. We retrospectively reviewed the medical charts of all patients evaluated between January 2017 and December 2019 in our adult neuropsychiatric genetics clinic at the McGill University Health Centre (MUHC), who had undergone a comprehensive ID/ASD gene panel. Thirty-four patients aged > 16 years, affected by ID/ASD and/or other neuropsychiatric/behavioral disorders, were identified. Pathogenic or likely pathogenic variants were identified in one-third of our cohort (32%): 8 single-nucleotide variants in 8 genes (CASK, SHANK3, IQSEC2, CHD2, ZBTB20, TREX1, SON, and TUBB2A) and 3 copy number variants (17p13.3, 16p13.12p13.11, and 9p24.3p24.1). The presence of psychiatric/behavioral disorders, regardless of the co-occurrence of ID, and, at a borderline level, the presence of ID alone were associated with positive genetic findings (p = 0.024 and p = 0.054, respectively). Moreover, seizures were associated with positive genetic results (p = 0.024). One-third of individuals presenting with psychiatric illness who met our red flags for Mendelian diseases have pathogenic or likely pathogenic variants which can be identified using a comprehensive ID/ASD gene panel (~ 2500 genes) performed on an exome backbone.
Collapse
Affiliation(s)
- Y Trakadis
- Division of Medical Genetics, Department of Specialized Medicine, Montreal Children's Hospital, McGill University Health Centre (MUHC), Room A04.3140, 1001 Boul. Décarie, Montreal, QC, H4A 3J1, Canada. .,Department of Human Genetics, McGill University, Montreal, QC, Canada. .,Douglas Mental Health Institute/Hospital, Montreal, Canada. .,Department of Psychiatry, McGill University, Montreal, Canada.
| | - A Accogli
- Division of Medical Genetics, Department of Specialized Medicine, Montreal Children's Hospital, McGill University Health Centre (MUHC), Room A04.3140, 1001 Boul. Décarie, Montreal, QC, H4A 3J1, Canada
| | - B Qi
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - D Bloom
- Douglas Mental Health Institute/Hospital, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada
| | - R Joober
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Douglas Mental Health Institute/Hospital, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada
| | - E Levy
- Douglas Mental Health Institute/Hospital, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada
| | - K Tabbane
- Douglas Mental Health Institute/Hospital, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada
| |
Collapse
|
18
|
Juvale IIA, Che Has AT. The Potential Role of miRNAs as Predictive Biomarkers in Neurodevelopmental Disorders. J Mol Neurosci 2021; 71:1338-1355. [PMID: 33774758 DOI: 10.1007/s12031-021-01825-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022]
Abstract
Neurodevelopmental disorders are defined as a set of abnormal brain developmental conditions marked by the early childhood onset of cognitive, behavioral, and functional deficits leading to memory and learning problems, emotional instability, and impulsivity. Autism spectrum disorder, attention-deficit/hyperactivity disorder, Tourette syndrome, fragile X syndrome, and Down's syndrome are a few known examples of neurodevelopmental disorders. Although they are relatively common in both developed and developing countries, very little is currently known about their underlying molecular mechanisms. Both genetic and environmental factors are known to increase the risk of neurodevelopmental disorders. Current diagnostic and screening tests for neurodevelopmental disorders are not reliable; hence, individuals with neurodevelopmental disorders are often diagnosed in the later stages. This negatively affects their prognosis and quality of life, prompting the need for a better diagnostic biomarker. Recent studies on microRNAs and their altered regulation in diseases have shed some light on the possible role they could play in the development of the central nervous system. This review attempts to elucidate our current understanding of the role that microRNAs play in neurodevelopmental disorders with the hope of utilizing them as potential biomarkers in the future.
Collapse
Affiliation(s)
- Iman Imtiyaz Ahmed Juvale
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Tarmizi Che Has
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
19
|
Ripamonti S, Shomroni O, Rhee JS, Chowdhury K, Jahn O, Hellmann KP, Bonn S, Brose N, Tirard M. SUMOylation controls the neurodevelopmental function of the transcription factor Zbtb20. J Neurochem 2020; 154:647-661. [PMID: 32233089 DOI: 10.1111/jnc.15008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
SUMOylation is a dynamic post-translational protein modification that primarily takes place in cell nuclei, where it plays a key role in multiple DNA-related processes. In neurons, the SUMOylation-dependent control of a subset of neuronal transcription factors is known to regulate various aspects of nerve cell differentiation, development, and function. In an unbiased screen for endogenous SUMOylation targets in the developing mouse brain, based on a His6 -HA-SUMO1 knock-in mouse line, we previously identified the transcription factor Zinc finger and BTB domain-containing 20 (Zbtb20) as a new SUMO1-conjugate. We show here that the three key SUMO paralogues SUMO1, SUMO2, and SUMO3 can all be conjugated to Zbtb20 in vitro in HEK293FT cells, and we confirm the SUMOylation of Zbtb20 in vivo in mouse brain. Using primary hippocampal neurons from wild-type and Zbtb20 knock-out (KO) mice as a model system, we then demonstrate that the expression of Zbtb20 is required for proper nerve cell development and neurite growth and branching. Furthermore, we show that the SUMOylation of Zbtb20 is essential for its function in this context, and provide evidence indicating that SUMOylation affects the Zbtb20-dependent transcriptional profile of neurons. Our data highlight the role of SUMOylation in the regulation of neuronal transcription factors that determine nerve cell development, and they demonstrate that key functions of the transcription factor Zbtb20 in neuronal development and neurite growth are under obligatory SUMOylation control.
Collapse
Affiliation(s)
- Silvia Ripamonti
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Orr Shomroni
- NGS Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Jeong Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kamal Chowdhury
- Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus Peter Hellmann
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Marilyn Tirard
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
20
|
Al-Naama N, Mackeh R, Kino T. C 2H 2-Type Zinc Finger Proteins in Brain Development, Neurodevelopmental, and Other Neuropsychiatric Disorders: Systematic Literature-Based Analysis. Front Neurol 2020; 11:32. [PMID: 32117005 PMCID: PMC7034409 DOI: 10.3389/fneur.2020.00032] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are multifaceted pathologic conditions manifested with intellectual disability, autistic features, psychiatric problems, motor dysfunction, and/or genetic/chromosomal abnormalities. They are associated with skewed neurogenesis and brain development, in part through dysfunction of the neural stem cells (NSCs) where abnormal transcriptional regulation on key genes play significant roles. Recent accumulated evidence highlights C2H2-type zinc finger proteins (C2H2-ZNFs), the largest transcription factor family in humans, as important targets for the pathologic processes associated with NDDs. In this review, we identified their significant accumulation (74 C2H2-ZNFs: ~10% of all human member proteins) in brain physiology and pathology. Specifically, we discuss their physiologic contribution to brain development, particularly focusing on their actions in NSCs. We then explain their pathologic implications in various forms of NDDs, such as morphological brain abnormalities, intellectual disabilities, and psychiatric disorders. We found an important tendency that poly-ZNFs and KRAB-ZNFs tend to be involved in the diseases that compromise gross brain structure and human-specific higher-order functions, respectively. This may be consistent with their characteristic appearance in the course of species evolution and corresponding contribution to these brain activities.
Collapse
Affiliation(s)
- Njoud Al-Naama
- Laboratory of Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Rafah Mackeh
- Laboratory of Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Tomoshige Kino
- Laboratory of Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|