1
|
Beebe JL, Martin CR, Sanders AFP, Guidry J, Faheem F, Atallah J, Beaton EA. Short communication: catechol-O-methyltransferase allelic variation in relation to psychological and hormonal indices of stress in children and adolescents with chromosome 22q11.2 deletion syndrome (22q11.2DS). J Appl Genet 2024:10.1007/s13353-024-00898-5. [PMID: 39126435 DOI: 10.1007/s13353-024-00898-5] [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: 04/19/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024]
Abstract
Chromosome 22q11.2 deletion syndrome (22q11.2DS) is a developmental disorder with high rates of anxiety and psychosis. Catechol-O-methyltransferase (COMT) regulates epinephrine (E), norepinephrine (NE), and dopamine (DA) and is implicated in both anxiety and psychotic disorders. The aim of this study was to determine how COMT variation relates to psychological anxiety and associated stress physiology responsiveness to better understand symptom heterogeneity in people with 22q11.2DS. We examined COMT allelic variation in relation to anxiety and hypothalamic-pituitary-adrenocortical (HPA) and sympathetic-adrenomedullary (SAM) hormonal stress indicators in 30 children and adolescents with 22q11.2DS. Contrary to expectation, individuals with the higher activity COMTval allele had higher anxiety levels versus those with the low activity (COMTmet) allele (p = 0.021; Glass' Δ = 0.69). Anxiety was not correlated with salivary cortisol (CORT) or alpha-amylase (sAA) in either group. Groups did not differ in CORT levels (p = 0.58), but the COMTmet group had higher sAA (p = 0.026; Glass' Δ = 0.67, uncorrected) suggesting greater SAM reactivity but not HPA activity. This suggests that COMT allelic variation may contribute to differences in acute SAM but not slower HPA stress reactivity in those with 22q11.2DS.
Collapse
Affiliation(s)
- Jessie L Beebe
- Department of Biology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA
| | - Cydney R Martin
- Department of Biology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA
| | - Ashley F P Sanders
- Department of Psychology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA
| | - Jeremy Guidry
- Department of Biology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA
| | - Fahad Faheem
- Department of Biology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA
| | - Joel Atallah
- Department of Biology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA.
| | - Elliott A Beaton
- Department of Psychology, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA, 70148, USA.
| |
Collapse
|
2
|
Cipriano A, Colantoni A, Calicchio A, Fiorentino J, Gomes D, Moqri M, Parker A, Rasouli S, Caldwell M, Briganti F, Roncarolo MG, Baldini A, Weinacht KG, Tartaglia GG, Sebastiano V. Transcriptional and epigenetic characterization of a new in vitro platform to model the formation of human pharyngeal endoderm. Genome Biol 2024; 25:211. [PMID: 39118163 PMCID: PMC11312149 DOI: 10.1186/s13059-024-03354-z] [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: 01/18/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND The Pharyngeal Endoderm (PE) is an extremely relevant developmental tissue, serving as the progenitor for the esophagus, parathyroids, thyroids, lungs, and thymus. While several studies have highlighted the importance of PE cells, a detailed transcriptional and epigenetic characterization of this important developmental stage is still missing, especially in humans, due to technical and ethical constraints pertaining to its early formation. RESULTS Here we fill this knowledge gap by developing an in vitro protocol for the derivation of PE-like cells from human Embryonic Stem Cells (hESCs) and by providing an integrated multi-omics characterization. Our PE-like cells robustly express PE markers and are transcriptionally homogenous and similar to in vivo mouse PE cells. In addition, we define their epigenetic landscape and dynamic changes in response to Retinoic Acid by combining ATAC-Seq and ChIP-Seq of histone modifications. The integration of multiple high-throughput datasets leads to the identification of new putative regulatory regions and to the inference of a Retinoic Acid-centered transcription factor network orchestrating the development of PE-like cells. CONCLUSIONS By combining hESCs differentiation with computational genomics, our work reveals the epigenetic dynamics that occur during human PE differentiation, providing a solid resource and foundation for research focused on the development of PE derivatives and the modeling of their developmental defects in genetic syndromes.
Collapse
Affiliation(s)
- Andrea Cipriano
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Alessio Colantoni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano Di Tecnologia (IIT), 00161, Rome, Italy
| | - Alessandro Calicchio
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Jonathan Fiorentino
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano Di Tecnologia (IIT), 00161, Rome, Italy
| | - Danielle Gomes
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Mahdi Moqri
- Biomedical Informatics Program, Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Alexander Parker
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Sajede Rasouli
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Matthew Caldwell
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Francesca Briganti
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Maria Grazia Roncarolo
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, 94305, USA
- Center for Definitive and Curative Medicine (CDCM), Stanford School of Medicine, Stanford, CA, USA
| | - Antonio Baldini
- Department of Molecular Medicine and Medical Biotech., University Federico II, 80131, Naples, Italy
| | - Katja G Weinacht
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Gian Gaetano Tartaglia
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano Di Tecnologia (IIT), 00161, Rome, Italy.
- Center for Human Technology, Fondazione Istituto Italiano Di Tecnologia (IIT), 16152, Genoa, Italy.
| | - Vittorio Sebastiano
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA, 94305, USA.
- Institute for Stem Cell Biology and Regenerative Medicine (ISCBRM), Stanford School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
3
|
Siraj L, Castro RI, Dewey H, Kales S, Nguyen TTL, Kanai M, Berenzy D, Mouri K, Wang QS, McCaw ZR, Gosai SJ, Aguet F, Cui R, Vockley CM, Lareau CA, Okada Y, Gusev A, Jones TR, Lander ES, Sabeti PC, Finucane HK, Reilly SK, Ulirsch JC, Tewhey R. Functional dissection of complex and molecular trait variants at single nucleotide resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592437. [PMID: 38766054 PMCID: PMC11100724 DOI: 10.1101/2024.05.05.592437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Identifying the causal variants and mechanisms that drive complex traits and diseases remains a core problem in human genetics. The majority of these variants have individually weak effects and lie in non-coding gene-regulatory elements where we lack a complete understanding of how single nucleotide alterations modulate transcriptional processes to affect human phenotypes. To address this, we measured the activity of 221,412 trait-associated variants that had been statistically fine-mapped using a Massively Parallel Reporter Assay (MPRA) in 5 diverse cell-types. We show that MPRA is able to discriminate between likely causal variants and controls, identifying 12,025 regulatory variants with high precision. Although the effects of these variants largely agree with orthogonal measures of function, only 69% can plausibly be explained by the disruption of a known transcription factor (TF) binding motif. We dissect the mechanisms of 136 variants using saturation mutagenesis and assign impacted TFs for 91% of variants without a clear canonical mechanism. Finally, we provide evidence that epistasis is prevalent for variants in close proximity and identify multiple functional variants on the same haplotype at a small, but important, subset of trait-associated loci. Overall, our study provides a systematic functional characterization of likely causal common variants underlying complex and molecular human traits, enabling new insights into the regulatory grammar underlying disease risk.
Collapse
Affiliation(s)
- Layla Siraj
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Biophysics, Harvard Graduate School of Arts and Sciences, Boston, MA, USA
- Harvard-Massachusetts Institute of Technology MD/PhD Program, Harvard Medical School, Boston, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | - Masahiro Kanai
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Qingbo S. Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | - Sager J. Gosai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - François Aguet
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Illumina Artificial Intelligence Laboratory, Illumina, San Diego, CA, USA
| | - Ran Cui
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Caleb A. Lareau
- Program in Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Alexander Gusev
- Harvard Medical School and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thouis R. Jones
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric S. Lander
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biology, MIT, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Pardis C. Sabeti
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hilary K. Finucane
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Steven K. Reilly
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Jacob C. Ulirsch
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
- Illumina Artificial Intelligence Laboratory, Illumina, San Diego, CA, USA
| | - Ryan Tewhey
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
4
|
von Scheibler ENMM, Widdershoven JCC, van Barneveld DCPBM, Schröder N, van Eeghen AM, van Amelsvoort TAMJ, Boot E. Hearing loss and history of otolaryngological conditions in adults with microdeletion 22q11.2. Am J Med Genet A 2024; 194:e63456. [PMID: 37916923 DOI: 10.1002/ajmg.a.63456] [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: 07/28/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
Previous studies have shown that the 22q11.2 microdeletion, associated with 22q11.2 deletion syndrome (22q11.2DS), conveys an increased risk of chronic otitis media, and hearing loss at young age. This study reports on hearing loss and history of otolaryngological conditions in adults with 22q11.2DS. We conducted a retrospective study of 60 adults with 22q11.2DS (41.7% male) at median age 25 (range 16-74) years who had visited an otolaryngologist and audiologist for routine assessment at a 22q11.2 expert center. Demographic, genetic, audiometric, and otolaryngological data were systematically extracted from the medical files. Regression analysis was used to evaluate the effect of age, sex, full-scale intelligence quotient, and history of chronic otitis media on the severity of hearing loss. Hearing loss, mostly high-frequency sensorineural, was found in 78.3% of adults. Higher age and history of chronic otitis media were associated with more severe hearing loss. Otolaryngological conditions with possible treatment implications included chronic otitis media (56.7%), globus pharyngeus (18.3%), balance problems (16.7%), and obstructive sleep apnea (8.3%). The results suggest that in 22q11.2DS, high-frequency hearing loss appears to be common from a young adult age, and often unrecognized. Therefore, we recommend periodic audiometric screening in all adults, including high-frequency ranges.
Collapse
Affiliation(s)
- Emma N M M von Scheibler
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands
| | - Josine C C Widdershoven
- Department of Otorhinolaryngology, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Nina Schröder
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands
| | - Agnies M van Eeghen
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands
- Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Erik Boot
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands
- The Dalglish Family 22q Clinic, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Antkowiak KR, Coskun P, Noronha ST, Tavella D, Massi F, Ryder SP. A nematode model to evaluate microdeletion phenotype expression. G3 (BETHESDA, MD.) 2024; 14:jkad258. [PMID: 37956108 PMCID: PMC10849325 DOI: 10.1093/g3journal/jkad258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/12/2023] [Accepted: 11/05/2023] [Indexed: 11/15/2023]
Abstract
Microdeletion syndromes are genetic diseases caused by multilocus chromosomal deletions too small to be detected by karyotyping. They are typified by complex pleiotropic developmental phenotypes that depend both on the extent of the deletion and variations in genetic background. Microdeletion alleles cause a wide array of consequences involving multiple pathways. How simultaneous haploinsufficiency of numerous adjacent genes leads to complex and variable pleiotropic phenotypes is not well understood. CRISPR/Cas9 genome editing has been shown to induce microdeletion-like alleles at a meaningful rate. Here, we describe a microdeletion allele in Caenorhabditis elegans recovered during a CRISPR/Cas9 genome editing experiment. We mapped the allele to chromosome V, balanced it with a reciprocal translocation crossover suppressor, and precisely defined the breakpoint junction. The allele simultaneously removes 32 protein-coding genes, yet animals homozygous for this mutation are viable as adults. Homozygous animals display a complex phenotype including maternal effect lethality, producing polynucleated embryos that grow into uterine tumors, vulva morphogenesis defects, body wall distensions, uncoordinated movement, and a shortened life span typified by death by bursting. Our work provides an opportunity to explore the complexity and penetrance of microdeletion phenotypes in a simple genetic model system.
Collapse
Affiliation(s)
- Katianna R Antkowiak
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Peren Coskun
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sharon T Noronha
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Davide Tavella
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Francesca Massi
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sean P Ryder
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
6
|
Perrottelli A, Marzocchi FF, Caporusso E, Giordano GM, Giuliani L, Melillo A, Pezzella P, Bucci P, Mucci A, Galderisi S. Advances in the understanding of the pathophysiology of schizophrenia and bipolar disorder through induced pluripotent stem cell models. J Psychiatry Neurosci 2024; 49:E109-E125. [PMID: 38490647 PMCID: PMC10950363 DOI: 10.1503/jpn.230112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/04/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024] Open
Abstract
The pathophysiology of schizophrenia and bipolar disorder involves a complex interaction between genetic and environmental factors that begins in the early stages of neurodevelopment. Recent advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising tool for understanding the neurobiological alterations involved in these disorders and, potentially, for developing new treatment options. In this review, we summarize the results of iPSC-based research on schizophrenia and bipolar disorder, showing disturbances in neurodevelopmental processes, imbalance in glutamatergic-GABAergic transmission and neuromorphological alterations. The limitations of the reviewed literature are also highlighted, particularly the methodological heterogeneity of the studies, the limited number of studies developing iPSC models of both diseases simultaneously, and the lack of in-depth clinical characterization of the included samples. Further studies are needed to advance knowledge on the common and disease-specific pathophysiological features of schizophrenia and bipolar disorder and to promote the development of new treatment options.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Giuliani
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Melillo
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Paola Bucci
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Armida Mucci
- From the University of Campania "Luigi Vanvitelli", Naples, Italy
| | | |
Collapse
|
7
|
Shin W, Kutmon M, Mina E, van Amelsvoort T, Evelo CT, Ehrhart F. Exploring pathway interactions to detect molecular mechanisms of disease: 22q11.2 deletion syndrome. Orphanet J Rare Dis 2023; 18:335. [PMID: 37872602 PMCID: PMC10594698 DOI: 10.1186/s13023-023-02953-6] [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: 09/22/2022] [Accepted: 10/10/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND 22q11.2 Deletion Syndrome (22q11DS) is a genetic disorder characterized by the deletion of adjacent genes at a location specified as q11.2 of chromosome 22, resulting in an array of clinical phenotypes including autistic spectrum disorder, schizophrenia, congenital heart defects, and immune deficiency. Many characteristics of the disorder are known, such as the phenotypic variability of the disease and the biological processes associated with it; however, the exact and systemic molecular mechanisms between the deleted area and its resulting clinical phenotypic expression, for example that of neuropsychiatric diseases, are not yet fully understood. RESULTS Using previously published transcriptomics data (GEO:GSE59216), we constructed two datasets: one set compares 22q11DS patients experiencing neuropsychiatric diseases versus healthy controls, and the other set 22q11DS patients without neuropsychiatric diseases versus healthy controls. We modified and applied the pathway interaction method, originally proposed by Kelder et al. (2011), on a network created using the WikiPathways pathway repository and the STRING protein-protein interaction database. We identified genes and biological processes that were exclusively associated with the development of neuropsychiatric diseases among the 22q11DS patients. Compared with the 22q11DS patients without neuropsychiatric diseases, patients experiencing neuropsychiatric diseases showed significant overrepresentation of regulated genes involving the natural killer cell function and the PI3K/Akt signalling pathway, with affected genes being closely associated with downregulation of CRK like proto-oncogene adaptor protein. Both the pathway interaction and the pathway overrepresentation analysis observed the disruption of the same biological processes, even though the exact lists of genes collected by the two methods were different. CONCLUSIONS Using the pathway interaction method, we were able to detect a molecular network that could possibly explain the development of neuropsychiatric diseases among the 22q11DS patients. This way, our method was able to complement the pathway overrepresentation analysis, by filling the knowledge gaps on how the affected pathways are linked to the original deletion on chromosome 22. We expect our pathway interaction method could be used for problems with similar contexts, where complex genetic mechanisms need to be identified to explain the resulting phenotypic plasticity.
Collapse
Affiliation(s)
- Woosub Shin
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Martina Kutmon
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - Eleni Mina
- Leiden University, Leiden, The Netherlands
| | | | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6229 ER, The Netherlands.
- Psychiatry & Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
8
|
Mancini V, Saleh MG, Delavari F, Bagautdinova J, Eliez S. Excitatory/Inhibitory Imbalance Underlies Hippocampal Atrophy in Individuals With 22q11.2 Deletion Syndrome With Psychotic Symptoms. Biol Psychiatry 2023; 94:569-579. [PMID: 37011759 DOI: 10.1016/j.biopsych.2023.03.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/10/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Abnormal neurotransmitter levels have been reported in individuals at high risk for schizophrenia, leading to a shift in the excitatory/inhibitory balance. However, it is unclear whether these alterations predate the onset of clinically relevant symptoms. Our aim was to explore in vivo measures of excitatory/inhibitory balance in 22q11.2 deletion carriers, a population at genetic risk for psychosis. METHODS Glx (glutamate+glutamine) and GABA+ (gamma-aminobutyric acid with macromolecules and homocarnosine) concentrations were estimated in the anterior cingulate cortex, superior temporal cortex, and hippocampus using the Mescher-Garwood point-resolved spectroscopy (MEGA-PRESS) sequence and the Gannet toolbox in 52 deletion carriers and 42 control participants. T1-weighted images were acquired longitudinally and processed with FreeSurfer version 6 to extract hippocampal volume. Subgroup analyses were conducted in deletion carriers with psychotic symptoms. RESULTS While no differences were found in the anterior cingulate cortex, deletion carriers had higher levels of Glx in the hippocampus and superior temporal cortex and lower levels of GABA+ in the hippocampus than control participants. We additionally found a higher Glx concentration in the hippocampus of deletion carriers with psychotic symptoms. Finally, more pronounced hippocampal atrophy was significantly associated with increased Glx levels in deletion carriers. CONCLUSIONS We provide evidence for an excitatory/inhibitory imbalance in temporal brain structures of deletion carriers, with a further hippocampal Glx increase in individuals with psychotic symptoms that was associated with hippocampal atrophy. These results are in line with theories proposing abnormally enhanced glutamate levels as a mechanistic explanation for hippocampal atrophy via excitotoxicity. Our results highlight a central role of glutamate in the hippocampus of individuals at genetic risk for schizophrenia.
Collapse
Affiliation(s)
- Valentina Mancini
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland.
| | - Muhammad G Saleh
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Farnaz Delavari
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Joëlle Bagautdinova
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Department of Genetic Medicine and Development, University of Geneva School of Medicine, Geneva, Switzerland
| |
Collapse
|
9
|
Grent-'t-Jong T, Brickwedde M, Metzner C, Uhlhaas PJ. 40-Hz Auditory Steady-State Responses in Schizophrenia: Toward a Mechanistic Biomarker for Circuit Dysfunctions and Early Detection and Diagnosis. Biol Psychiatry 2023; 94:550-560. [PMID: 37086914 DOI: 10.1016/j.biopsych.2023.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/24/2023]
Abstract
There is converging evidence that 40-Hz auditory steady-state responses (ASSRs) are robustly impaired in schizophrenia and could constitute a potential biomarker for characterizing circuit dysfunctions as well as enable early detection and diagnosis. Here, we provide an overview of the mechanisms involved in 40-Hz ASSRs, drawing on computational, physiological, and pharmacological data with a focus on parameters modulating the balance between excitation and inhibition. We will then summarize findings from electro- and magnetoencephalographic studies in participants at clinical high risk for psychosis, patients with first-episode psychosis, and patients with schizophrenia to identify the pattern of deficits across illness stages, the relationship with clinical variables, and the prognostic potential. Finally, data on genetics and developmental modifications will be reviewed, highlighting the importance of late modifications of 40-Hz ASSRs during adolescence, which are closely related to the underlying changes in GABA (gamma-aminobutyric acid) interneurons. Together, our review suggests that 40-Hz ASSRs may constitute an informative electrophysiological approach to characterize circuit dysfunctions in psychosis that could be relevant for the development of mechanistic biomarkers.
Collapse
Affiliation(s)
- Tineke Grent-'t-Jong
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marion Brickwedde
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Metzner
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany; School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom
| | - Peter J Uhlhaas
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
10
|
Campbell PD, Lee I, Thyme S, Granato M. Mitochondrial proteins encoded by the 22q11.2 neurodevelopmental locus regulate neural stem and progenitor cell proliferation. Mol Psychiatry 2023; 28:3769-3781. [PMID: 37794116 PMCID: PMC10730408 DOI: 10.1038/s41380-023-02272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023]
Abstract
Microdeletion of a 3Mb region encompassing 45 protein-coding genes at chromosome 22q11.2 (22q11.2DS) predisposes individuals to multiple neurodevelopmental disorders and is one of the greatest genetic risk factors for schizophrenia. Defective mitochondrial function has been hypothesized to contribute to 22q11.2DS pathogenesis; however, which of the six mitochondrial genes contribute to neurodevelopmental phenotypes and their underlying mechanisms remain unresolved. To systematically test 22q11.2DS genes for functional roles in neurodevelopment and behavior, we generated genetic mutants for each of the 37 conserved zebrafish orthologs and performed high throughput behavioral phenotyping using seven behavioral assays. Through this unbiased approach, we identified five single-gene mutants with partially overlapping behavioral phenotypes. Two of these genes, mrpl40 and prodha, encode for mitochondrial proteins and, similar to what we observed in mrpl40 and prodha mutants, pharmacologic inhibition of mitochondrial function during development results in microcephaly. Single mutant analysis shows that both mrpl40 and prodha mutants display aberrant neural stem and progenitor cell proliferation, with each gene regulating distinct cell populations. Finally, double mutants for both mrpl40 and prodha display aggravated behavioral phenotypes and neural stem and progenitor cell analysis reveals a previously unrecognized partially redundant role for mrpl40 and prodha in regulating radial glia-like cell proliferation. Combined, our results demonstrate a critical role for mitochondrial function in neural stem and progenitor cell populations in the developing vertebrate brain and provide compelling evidence that mitochondrial dysfunction during neurodevelopment is linked to brain volume and behavioral phenotypes observed in models of 22q11.2DS.
Collapse
Affiliation(s)
- Philip D Campbell
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Isaiah Lee
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Summer Thyme
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Rukh S, Meechan DW, Maynard TM, Lamantia AS. Out of Line or Altered States? Neural Progenitors as a Target in a Polygenic Neurodevelopmental Disorder. Dev Neurosci 2023; 46:1-21. [PMID: 37231803 DOI: 10.1159/000530898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
The genesis of a mature complement of neurons is thought to require, at least in part, precursor cell lineages in which neural progenitors have distinct identities recognized by exclusive expression of one or a few molecular markers. Nevertheless, limited progenitor types distinguished by specific markers and lineal progression through such subclasses cannot easily yield the magnitude of neuronal diversity in most regions of the nervous system. The late Verne Caviness, to whom this edition of Developmental Neuroscience is dedicated, recognized this mismatch. In his pioneering work on the histogenesis of the cerebral cortex, he acknowledged the additional flexibility required to generate multiple classes of cortical projection and interneurons. This flexibility may be accomplished by establishing cell states in which levels rather than binary expression or repression of individual genes vary across each progenitor's shared transcriptome. Such states may reflect local, stochastic signaling via soluble factors or coincidence of cell surface ligand/receptor pairs in subsets of neighboring progenitors. This probabilistic, rather than determined, signaling could modify transcription levels via multiple pathways within an apparently uniform population of progenitors. Progenitor states, therefore, rather than lineal relationships between types may underlie the generation of neuronal diversity in most regions of the nervous system. Moreover, mechanisms that influence variation required for flexible progenitor states may be targets for pathological changes in a broad range of neurodevelopmental disorders, especially those with polygenic origins.
Collapse
Affiliation(s)
- Shah Rukh
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Daniel W Meechan
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Thomas M Maynard
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Anthony-Samuel Lamantia
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
12
|
Szczawińska-Popłonyk A, Schwartzmann E, Chmara Z, Głukowska A, Krysa T, Majchrzycki M, Olejnicki M, Ostrowska P, Babik J. Chromosome 22q11.2 Deletion Syndrome: A Comprehensive Review of Molecular Genetics in the Context of Multidisciplinary Clinical Approach. Int J Mol Sci 2023; 24:ijms24098317. [PMID: 37176024 PMCID: PMC10179617 DOI: 10.3390/ijms24098317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The 22q11.2 deletion syndrome is a multisystemic disorder characterized by a marked variability of phenotypic features, making the diagnosis challenging for clinicians. The wide spectrum of clinical manifestations includes congenital heart defects-most frequently conotruncal cardiac anomalies-thymic hypoplasia and predominating cellular immune deficiency, laryngeal developmental defects, midline anomalies with cleft palate and velar insufficiency, structural airway defects, facial dysmorphism, parathyroid and thyroid gland hormonal dysfunctions, speech delay, developmental delay, and neurocognitive and psychiatric disorders. Significant progress has been made in understanding the complex molecular genetic etiology of 22q11.2 deletion syndrome underpinning the heterogeneity of clinical manifestations. The deletion is caused by chromosomal rearrangements in meiosis and is mediated by non-allelic homologous recombination events between low copy repeats or segmental duplications in the 22q11.2 region. A range of genetic modifiers and environmental factors, as well as the impact of hemizygosity on the remaining allele, contribute to the intricate genotype-phenotype relationships. This comprehensive review has been aimed at highlighting the molecular genetic background of 22q11.2 deletion syndrome in correlation with a clinical multidisciplinary approach.
Collapse
Affiliation(s)
- Aleksandra Szczawińska-Popłonyk
- Department of Pediatric Pneumonology, Allergy and Clinical Immunology, Institute of Pediatrics, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Eyal Schwartzmann
- Medical Student Scientific Society, English Division, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Zuzanna Chmara
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Antonina Głukowska
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Tomasz Krysa
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Maksymilian Majchrzycki
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Maurycy Olejnicki
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Paulina Ostrowska
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Joanna Babik
- Gynecology and Obstetrics with Pregnancy Pathology Unit, Franciszek Raszeja Municipal Hospital, 60-834 Poznań, Poland
| |
Collapse
|
13
|
Kocere A, Lalonde RL, Mosimann C, Burger A. Lateral thinking in syndromic congenital cardiovascular disease. Dis Model Mech 2023; 16:dmm049735. [PMID: 37125615 PMCID: PMC10184679 DOI: 10.1242/dmm.049735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Syndromic birth defects are rare diseases that can present with seemingly pleiotropic comorbidities. Prime examples are rare congenital heart and cardiovascular anomalies that can be accompanied by forelimb defects, kidney disorders and more. Whether such multi-organ defects share a developmental link remains a key question with relevance to the diagnosis, therapeutic intervention and long-term care of affected patients. The heart, endothelial and blood lineages develop together from the lateral plate mesoderm (LPM), which also harbors the progenitor cells for limb connective tissue, kidneys, mesothelia and smooth muscle. This developmental plasticity of the LPM, which founds on multi-lineage progenitor cells and shared transcription factor expression across different descendant lineages, has the potential to explain the seemingly disparate syndromic defects in rare congenital diseases. Combining patient genome-sequencing data with model organism studies has already provided a wealth of insights into complex LPM-associated birth defects, such as heart-hand syndromes. Here, we summarize developmental and known disease-causing mechanisms in early LPM patterning, address how defects in these processes drive multi-organ comorbidities, and outline how several cardiovascular and hematopoietic birth defects with complex comorbidities may be LPM-associated diseases. We also discuss strategies to integrate patient sequencing, data-aggregating resources and model organism studies to mechanistically decode congenital defects, including potentially LPM-associated orphan diseases. Eventually, linking complex congenital phenotypes to a common LPM origin provides a framework to discover developmental mechanisms and to anticipate comorbidities in congenital diseases affecting the cardiovascular system and beyond.
Collapse
Affiliation(s)
- Agnese Kocere
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
- Department of Molecular Life Science, University of Zurich, 8057 Zurich, Switzerland
| | - Robert L. Lalonde
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Alexa Burger
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| |
Collapse
|
14
|
Zhu C, Yang Y, Pan B, Wei H, Ju J, Si N, Xu Q. Genetic Screening of Targeted Region on the Chromosome 22q11.2 in Patients with Microtia and Congenital Heart Defect. Genes (Basel) 2023; 14:genes14040879. [PMID: 37107637 PMCID: PMC10137977 DOI: 10.3390/genes14040879] [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: 02/10/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Microtia is a congenital malformation characterized by a small, abnormally shaped auricle (pinna) ranging in severity. Congenital heart defect (CHD) is one of the comorbid anomalies with microtia. However, the genetic basis of the co-existence of microtia and CHD remains unclear. Copy number variations (CNVs) of 22q11.2 contribute significantly to microtia and CHD, respectively, thus suggesting a possible shared genetic cause embedded in this genomic region. In this study, 19 sporadic patients with microtia and CHD, as well as a nuclear family, were enrolled for genetic screening of single nucleotide variations (SNVs) and CNVs in 22q11.2 by target capture sequencing. We detected a total of 105 potential deleterious variations, which were enriched in ear- or heart-development-related genes, including TBX1 and DGCR8. The gene burden analysis also suggested that these genes carry more deleterious mutations in the patients, as well as several other genes associated with cardiac development, such as CLTCL1. Additionally, a microduplication harboring SUSD2 was validated in an independent cohort. This study provides new insights into the underlying mechanisms for the comorbidity of microtia and CHD focusing on chromosome 22q11.2, and suggests that a combination of genetic variations, including SNVs and CNVs, may play a crucial role instead of single gene mutation.
Collapse
Affiliation(s)
- Caiyun Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yang Yang
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100144, China
| | - Bo Pan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100144, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Jiahang Ju
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Nuo Si
- Research Center, Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100144, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
15
|
Neuroinflammation and Oxidative Stress in Individuals Affected by DiGeorge Syndrome. Int J Mol Sci 2023; 24:ijms24044242. [PMID: 36835652 PMCID: PMC9965448 DOI: 10.3390/ijms24044242] [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: 11/01/2022] [Revised: 02/07/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
DiGeorge syndrome (DGS) is a rare genetic disease caused by microdeletions of the 22q11.2 region (DGS1). A haploinsufficiency at 10p level has been proposed also as a DGS cause (DGS2). Clinical manifestations are variable. The most frequent features are thymic hypoplasia or aplasia with consequent immune deficiency, cardiac malformations, hypoparathyroidism, facial and palatine abnormalities, variable degrees of cognitive impairment and psychiatric disorders. The specific aim of this descriptive report is to discuss the correlation between oxidative stress and neuroinflammation in DGS patients with microdeletions of the 22q11.2 region. The deleted chromosomic region maps various genes involved in mitochondrial metabolisms, such as DGCR8 and TXNRD2, that could lead to reactive oxygen species (ROS) increased production and antioxidant depletion. Furthermore, increased levels of ROS in mitochondria would lead to the destruction of the projection neurons in the cerebral cortex with consequent neurocognitive impairment. Finally, the increase in modified protein belonging to the family of sulfoxide compounds and hexoses, acting as inhibitors of the IV and V mitochondria complex, could result in direct ROS overproduction. Neuroinflammation in DGS individuals could be directly related to the development of the syndrome's characteristic psychiatric and cognitive disorders. In patients with psychotic disorders, the most frequent psychiatric manifestation in DGS, Th-17, Th-1 and Th-2 cells are increased with consequent elevation of proinflammatory cytokine IL-6 and IL1β. In patients with anxiety disorders, both CD3 and CD4 are increased. Some patients with autism spectrum disorders (ASDs) have an augmented level of proinflammatory cytokines IL-12, IL-6 and IL-1β, while IFNγ and the anti-inflammatory cytokine IL-10 seem to be reduced. Other data proposed that altered synaptic plasticity could be directly involved in DGS cognitive disorders. In conclusion, the use of antioxidants for restoring mitochondrial functionality in DGS could be a useful tool to protect cortical connectivity and cognitive behavior.
Collapse
|
16
|
Floriani MA, Santos AS, Diniz BL, Glaeser AB, Gazzola Zen PR, Machado Rosa RF. 22q11 Copy Number Variations in a Brazilian Cohort of Children with Congenital Heart Disorders. Mol Syndromol 2023; 14:1-10. [PMID: 36777701 PMCID: PMC9911999 DOI: 10.1159/000525247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/19/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Congenital heart disease (CHD) is the most common type of congenital defect reported to be one of the leading causes of mortality in the first year of life. Microdeletion and microduplication syndromes (MMS) are associated with cardiac malformations. Understanding which genetic factors are involved in these conditions directly impacts treatment decisions. We aimed to identify the occurrence of genetic alterations and their association with MMS in CHD pediatric patients evaluated in a reference service of Southern Brazil. Methods Participants were recruited during 2010 in the intensive care unit of a pediatric hospital. MMs and regions of chromosome 22 were screened by SALSA MLPA Probemix P245 Microdeletion Syndromes-1A kit for detection of copy number variations (CNVs). Results MMS were detected in 11 from 207 patients (5.3%). Heterozygous deletion in the 22q11.2 chromosome region was the most prevalent CNV (5 from 11 patients). Also, atypical RTDR1 deletion and 22q11.2 duplication were detected. MLPA was able to reveal microdeletions in SNRPN and NF1 genes in patients with a normal karyotype and FISH. Conclusion Our study reports the prevalence and variability of genomic alterations associated with MMS in CHD pediatric patients. The results by MLPA are of great help in planning and specialized care.
Collapse
Affiliation(s)
- Maiara A. Floriani
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | - Bruna L. Diniz
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Andressa B. Glaeser
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Paulo R. Gazzola Zen
- Department of Internal Medicine, Clinical Genetics, UFCSPA, Porto Alegre, Brazil,Irmandade da Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, Brazil
| | - Rafael F. Machado Rosa
- Department of Internal Medicine, Clinical Genetics, UFCSPA, Porto Alegre, Brazil,Irmandade da Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, Brazil,*Rafael Fabiano Machado Rosa,
| |
Collapse
|
17
|
Campbell PD, Lee I, Thyme S, Granato M. Mitochondrial genes in the 22q11.2 deleted region regulate neural stem and progenitor cell proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522615. [PMID: 36711666 PMCID: PMC9881859 DOI: 10.1101/2023.01.03.522615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Microdeletion of a 3Mbp region encompassing 45 protein-coding genes at chromosome 22q11.2 (22q11.2DS) predisposes to multiple neurodevelopmental disorders and is one of the greatest genetic risk factors for schizophrenia. Defective mitochondrial function has been hypothesized to contribute to 22q11.2DS pathogenesis; however, which of the six mitochondrial genes contribute to neurodevelopmental phenotypes and their underlying mechanisms remain unresolved. To systematically test 22q11.2DS genes for functional roles in neurodevelopment and behavior, we generated genetic mutants for each of the 37 conserved zebrafish orthologs and performed high throughput behavioral phenotyping using seven behavioral assays. Through this unbiased approach, we identified five single-gene mutants with partially overlapping behavioral phenotypes. Two of these genes, mrpl40 and prodha , encode for mitochondrial proteins and, similar to what we observed in mrpl40 and prodha mutants, pharmacologic inhibition of mitochondrial function during development results in microcephaly. Finally, we show that both mrpl40 and prodha mutants display neural stem and progenitor cell phenotypes, with each gene regulating different neural stem cell populations. Combined, our results demonstrate a critical role for mitochondrial function in neural stem and progenitor cell populations in the developing vertebrate brain and provide compelling evidence that mitochondrial dysfunction during neurodevelopment is linked to brain volume and behavioral phenotypes observed in models of 22q11.2DS.
Collapse
Affiliation(s)
- Philip D. Campbell
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA, 19104
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA, 19104
| | - Isaiah Lee
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA, 19104
| | - Summer Thyme
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA, 35294
| | - Michael Granato
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA, 19104
| |
Collapse
|
18
|
Peter B. A case with cardiac, skeletal, speech, and motor traits narrows the subtelomeric 19p13.3 microdeletion region to 46 kb. Am J Med Genet A 2023; 191:120-129. [PMID: 36271830 DOI: 10.1002/ajmg.a.62998] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/22/2022] [Accepted: 10/07/2022] [Indexed: 12/14/2022]
Abstract
Subtelomeric 19p13.3 deletions have been associated with diverse anatomical and developmental phenotypes. A recent study of eight patients with subtelomeric interstitial 19p13.3 microdeletions at 0.3-1.4 Mb (hg 19) showed associations with growth restrictions, skeletal deformities, craniofacial anomalies, congenital heart defects, renal malformations, hernias, immune system deficits, fine and gross motor delays, speech delays, and developmental and learning delays. The authors defined two small regions of overlap containing four and 11 genes, respectively, with potential haploinsufficiency. Here, we present a new case with a de novo 184 kb deletion containing eight genes, three of which fall into the second previously identified small region of overlap, reducing the shared region to 46 kb. Phenotypic traits include most of the core findings in the previously reported cases but not growth restrictions, craniofacial anomalies, renal malformation, and learning disability. A closer look at the speech and motor delays reveals apraxic speech and discoordination in the fine and gross motor domain, consistent with cerebellar involvement across motor systems. Findings are consistent with a role of AZU1 in the observed immune deficiencies and PTBP1 in the observed skeletal, abdominal, speech, language, motor, and sensory traits. This case thus contributes to a more nuanced understanding of the subtelomeric 19p13.3 deletion region.
Collapse
Affiliation(s)
- Beate Peter
- College of Health Solutions, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
19
|
Montemurro N, Ricciardi L, Scerrati A, Ippolito G, Lofrese G, Trungu S, Stoccoro A. The Potential Role of Dysregulated miRNAs in Adolescent Idiopathic Scoliosis and 22q11.2 Deletion Syndrome. J Pers Med 2022; 12:1925. [PMID: 36422101 PMCID: PMC9695868 DOI: 10.3390/jpm12111925] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 08/29/2023] Open
Abstract
Background: Adolescent idiopathic scoliosis (AIS), affecting 2-4% of adolescents, is a multifactorial spinal disease. Interactions between genetic and environmental factors can influence disease onset through epigenetic mechanisms, including DNA methylation, histone modifications and miRNA expression. Recent evidence reported that, among all clinical features in individuals with 22q11.2 deletion syndrome (DS), scoliosis can occur with a higher incidence than in the general population. Methods: A PubMed and Ovid Medline search was performed for idiopathic scoliosis in the setting of 22q11.2DS and miRNA according to PRISMA guidelines. Results: Four papers, accounting for 2841 individuals, reported clinical data about scoliosis in individuals with 22q11.2DS, showing that approximately 35.1% of the individuals with 22q11.2DS developed scoliosis. Conclusions: 22q11.2DS could be used as a model for the study of AIS. The DGCR8 gene seems to be essential for microRNA biogenesis, which is why we propose that a possible common pathological mechanism between scoliosis and 22q11.2DS could be the dysregulation of microRNA expression. In the current study, we identified two miRNAs that were altered in both 22q11.2DS and AIS, miR-93 and miR-1306, thus, corroborating the hypothesis that the two diseases share common molecular alterations.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliera Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| | - Luca Ricciardi
- Department of NESMOS, Sapienza University of Rome, 00185 Roma, Italy
| | - Alba Scerrati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Ippolito
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), DSBMC Sapienza Università di Roma-Polo Pontino, 04100 Latina, Italy
| | - Giorgio Lofrese
- Division of Neurosurgery, Ospedale Bufalini, 47023 Cesena, Italy
| | - Sokol Trungu
- Department of NESMOS, Sapienza University of Rome, 00185 Roma, Italy
| | - Andrea Stoccoro
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
20
|
Mancini V, Rochas V, Seeber M, Grent-'t-Jong T, Rihs TA, Latrèche C, Uhlhaas PJ, Michel CM, Eliez S. Oscillatory Neural Signatures of Visual Perception Across Developmental Stages in Individuals With 22q11.2 Deletion Syndrome. Biol Psychiatry 2022; 92:407-418. [PMID: 35550793 DOI: 10.1016/j.biopsych.2022.02.961] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Numerous behavioral studies have highlighted the contribution of visual perceptual deficits to the nonverbal cognitive profile of individuals with 22q11.2 deletion syndrome. However, the neurobiological processes underlying these widespread behavioral alterations are yet to be fully understood. Thus, in this paper, we investigated the role of neural oscillations toward visuoperceptual deficits to elucidate the neurobiology of sensory impairments in deletion carriers. METHODS We acquired 125 high-density electroencephalography recordings during a visual grating task in a group of 62 deletion carriers and 63 control subjects. Stimulus-elicited oscillatory responses were analyzed with 1) time-frequency analysis using wavelets decomposition at sensor and source level, 2) intertrial phase coherence, and 3) Granger causality connectivity in source space. Additional analyses examined the development of neural oscillations across age bins. RESULTS Deletion carriers had decreased theta-band (4-8 Hz) and gamma-band (58-68 Hz) spectral power compared with control subjects in response to the visual stimuli, with an absence of age-related increase of theta- and gamma-band responses. Moreover, adult deletion carriers had decreased gamma- and theta-band responses but increased alpha/beta desynchronization (10-25 Hz) that correlated with behavioral performance. Granger causality estimates reflected an increased frontal-occipital connectivity in the beta range (22-40 Hz). CONCLUSIONS Deletion carriers exhibited decreased theta- and gamma-band responses to visual stimuli, while alpha/beta desynchronization was preserved. Overall, the lack of age-related changes in deletion carriers implicates developmental impairments in circuit mechanisms underlying neural oscillations. The dissociation between the maturation of theta/gamma- and alpha/beta-band responses may indicate a selective impairment in supragranular cortical layers, leading to compensatory top-down connectivity.
Collapse
Affiliation(s)
- Valentina Mancini
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland.
| | - Vincent Rochas
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland; Human Neuroscience Platform, Fondation Campus Biotech Geneva, Geneva, Switzerland
| | - Martin Seeber
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Tineke Grent-'t-Jong
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland; Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Tonia A Rihs
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Caren Latrèche
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Peter J Uhlhaas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland; Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Christoph M Michel
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland; Center for Biomedical Imaging, Lausanne, Switzerland
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Department of Genetic Medicine and Development, University of Geneva School of Medicine, Geneva, Switzerland
| |
Collapse
|
21
|
Abstract
The human brain consumes five orders of magnitude more energy than the sun by unit of mass and time. This staggering bioenergetic cost serves mostly synaptic transmission and actin cytoskeleton dynamics. The peak of both brain bioenergetic demands and the age of onset for neurodevelopmental disorders is approximately 5 years of age. This correlation suggests that defects in the machinery that provides cellular energy would be causative and/or consequence of neurodevelopmental disorders. We explore this hypothesis from the perspective of the machinery required for the synthesis of the electron transport chain, an ATP-producing and NADH-consuming enzymatic cascade. The electron transport chain is constituted by nuclear- and mitochondrial-genome-encoded subunits. These subunits are synthesized by the 80S and the 55S ribosomes, which are segregated to the cytoplasm and the mitochondrial matrix, correspondingly. Mitochondrial protein synthesis by the 55S ribosome is the rate-limiting step in the synthesis of electron transport chain components, suggesting that mitochondrial protein synthesis is a bottleneck for tissues with high bionergetic demands. We discuss genetic defects in the human nuclear and mitochondrial genomes that affect these protein synthesis machineries and cause a phenotypic spectrum spanning autism spectrum disorders to neurodegeneration during neurodevelopment. We propose that dysregulated mitochondrial protein synthesis is a chief, yet understudied, causative mechanism of neurodevelopmental and behavioral disorders.
Collapse
|
22
|
Takada I, Hidano S, Takahashi S, Yanaka K, Ogawa H, Tsuchiya M, Yokoyama A, Sato S, Ochi H, Nakagawa T, Kobayashi T, Nakagawa S, Makishima M. Transcriptional coregulator Ess2 controls survival of post-thymic CD4 + T cells through the Myc and IL-7 signaling pathways. J Biol Chem 2022; 298:102342. [PMID: 35933014 PMCID: PMC9436822 DOI: 10.1016/j.jbc.2022.102342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 11/21/2022] Open
Abstract
Ess2, also known as Dgcr14, is a transcriptional co-regulator of CD4+ T cells. Ess2 is located in a chromosomal region, the loss of which has been associated with 22q11.2 deletion syndrome (22q11DS), which causes heart defects, skeletal abnormalities, and immunodeficiency. However, the specific association of Ess2 with 22q11DS remains unclear. To elucidate the role of Ess2 in T-cell development, we generated Ess2 floxed (Ess2fl/fl) and CD4+ T cell-specific Ess2 KO (Ess2ΔCD4/ΔCD4) mice using the Cre/loxP system. Interestingly, Ess2ΔCD4/ΔCD4 mice exhibited reduced naïve T-cell numbers in the spleen, while the number of thymocytes (CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+) in the thymus remained unchanged. Furthermore, Ess2ΔCD4/ΔCD4 mice had decreased NKT cells and increased γδT cells in the thymus and spleen. A genome-wide expression analysis using RNA-seq revealed that Ess2 deletion alters the expression of many genes in CD4 single-positive thymocytes, including genes related to the immune system and Myc target genes. In addition, Ess2 enhanced the transcriptional activity of c-Myc. Some genes identified as Ess2 targets in mice show expressional correlation with ESS2 in human immune cells. Moreover, Ess2ΔCD4/ΔCD4 naïve CD4+ T cells did not maintain survival in response to IL-7. Our results suggest that Ess2 plays a critical role in post-thymic T-cell survival through the Myc and IL-7 signaling pathways.
Collapse
Affiliation(s)
- Ichiro Takada
- Division of Biochemistry, Department of Biomedical Sciences, School of Medicine, Nihon University, Itabashi-ku, Tokyo, Japan.
| | - Shinya Hidano
- Department of Infectious Diseases Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Sayuri Takahashi
- Department of Urology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kaori Yanaka
- RNA Biology Laboratory, RIKEN Advanced Research Institute, Wako, Saitama, Japan
| | - Hidesato Ogawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Megumi Tsuchiya
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Medical Hospital, Tokyo, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Tohru Nakagawa
- Department of Urology, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Takashi Kobayashi
- Department of Infectious Diseases Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, School of Medicine, Nihon University, Itabashi-ku, Tokyo, Japan.
| |
Collapse
|
23
|
Jima DD, Skaar DA, Planchart A, Motsinger-Reif A, Cevik SE, Park SS, Cowley M, Wright F, House J, Liu A, Jirtle RL, Hoyo C. Genomic map of candidate human imprint control regions: the imprintome. Epigenetics 2022; 17:1920-1943. [PMID: 35786392 PMCID: PMC9665137 DOI: 10.1080/15592294.2022.2091815] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Imprinted genes – critical for growth, metabolism, and neuronal function – are expressed from one parental allele. Parent-of-origin-dependent CpG methylation regulates this expression at imprint control regions (ICRs). Since ICRs are established before tissue specification, these methylation marks are similar across cell types. Thus, they are attractive for investigating the developmental origins of adult diseases using accessible tissues, but remain unknown. We determined genome-wide candidate ICRs in humans by performing whole-genome bisulphite sequencing (WGBS) of DNA derived from the three germ layers and from gametes. We identified 1,488 hemi-methylated candidate ICRs, including 19 of 25 previously characterized ICRs (https://humanicr.org/). Gamete methylation approached 0% or 100% in 332 ICRs (178 paternally and 154 maternally methylated), supporting parent-of-origin-specific methylation, and 65% were in well-described CTCF-binding or DNaseI hypersensitive regions. This draft of the human imprintome will allow for the systematic determination of the role of early-acquired imprinting dysregulation in the pathogenesis of human diseases and developmental and behavioural disorders.
Collapse
Affiliation(s)
- Dereje D Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - David A Skaar
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Antonio Planchart
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Alison Motsinger-Reif
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sebnem E Cevik
- Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Sarah S Park
- Toxicology Program, North Carolina State University, Raleigh, NC, USA.,Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Michael Cowley
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Fred Wright
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - John House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA.,National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Andy Liu
- Department of Neurology, Duke University, School of Medicine, Durham, NC, USA
| | - Randy L Jirtle
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Cathrine Hoyo
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
24
|
Epigenetics of Autism Spectrum Disorder: Histone Deacetylases. Biol Psychiatry 2022; 91:922-933. [PMID: 35120709 DOI: 10.1016/j.biopsych.2021.11.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 01/08/2023]
Abstract
The etiology of autism spectrum disorder (ASD) remains unknown, but gene-environment interactions, mediated through epigenetic mechanisms, are thought to be a key contributing factor. Prenatal environmental factors have been shown to be associated with both increased risk of ASD and altered histone deacetylases (HDACs) or acetylation levels. The relationship between epigenetic changes and gene expression in ASD suggests that alterations in histone acetylation, which lead to changes in gene transcription, may play a key role in ASD. Alterations in the acetylome have been demonstrated for several genes in ASD, including genes involved in synaptic function, neuronal excitability, and immune responses, which are mechanisms previously implicated in ASD. We review preclinical and clinical studies that investigated HDACs and autism-associated behaviors and discuss risk genes for ASD that code for proteins associated with HDACs. HDACs are also implicated in neurodevelopmental disorders with a known genetic etiology, such as 15q11-q13 duplication and Phelan-McDermid syndrome, which share clinical features and diagnostic comorbidities (e.g., epilepsy, anxiety, and intellectual disability) with ASD. Furthermore, we highlight factors that affect the behavioral phenotype of acetylome changes, including sensitive developmental periods and brain region specificity in the context of epigenetic programming.
Collapse
|
25
|
Mancini V, Rochas V, Seeber M, Roehri N, Rihs TA, Ferat V, Schneider M, Uhlhaas PJ, Eliez S, Michel CM. Aberrant Developmental Patterns of Gamma-Band Response and Long-Range Communication Disruption in Youths With 22q11.2 Deletion Syndrome. Am J Psychiatry 2022; 179:204-215. [PMID: 35236117 DOI: 10.1176/appi.ajp.2021.21020190] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Brain oscillations play a pivotal role in synchronizing responses of local and global ensembles of neurons. Patients with schizophrenia exhibit impairments in oscillatory response, which are thought to stem from abnormal maturation during critical developmental stages. Studying individuals at genetic risk for psychosis, such as 22q11.2 deletion carriers, from childhood to adulthood may provide insights into developmental abnormalities. METHODS The authors acquired 106 consecutive T1-weighted MR images and 40-Hz auditory steady-state responses (ASSRs) with high-density (256 channel) EEG in a group of 58 22q11.2 deletion carriers and 48 healthy control subjects. ASSRs were analyzed with 1) time-frequency analysis using Morlet wavelet decomposition, 2) intertrial phase coherence (ITPC), and 3) theta-gamma phase-amplitude coupling estimated in the source space between brain regions activated by the ASSRs. Additionally, volumetric analyses were performed with FreeSurfer. Subanalyses were conducted in deletion carriers who endorsed psychotic symptoms and in subgroups with different age bins. RESULTS Deletion carriers had decreased theta and late-latency 40-Hz ASSRs and phase synchronization compared with control subjects. Deletion carriers with psychotic symptoms displayed a further reduction of gamma-band response, decreased ITPC, and decreased top-down modulation of gamma-band response in the auditory cortex. Reduced gamma-band response was correlated with the atrophy of auditory cortex in individuals with psychotic symptoms. In addition, a linear increase of theta and gamma power from childhood to adulthood was found in control subjects but not in deletion carriers. CONCLUSIONS The results suggest that while all deletion carriers exhibit decreased gamma-band response, more severe local and long-range communication abnormalities are associated with the emergence of psychotic symptoms and gray matter loss. Additionally, the lack of age-related changes in deletion carriers indexes a potential developmental impairment in circuits underlying the maturation of neural oscillations during adolescence. The progressive disruption of gamma-band response in 22q11.2 deletion syndrome supports a developmental perspective toward understanding and treating psychotic disorders.
Collapse
Affiliation(s)
- Valentina Mancini
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Vincent Rochas
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Martin Seeber
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Nicolas Roehri
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Tonia A Rihs
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Victor Ferat
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Maude Schneider
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Peter J Uhlhaas
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| | - Christoph M Michel
- Developmental Imaging and Psychopathology Laboratory (Mancini, Schneider, Eliez) and Department of Genetic Medicine and Development (Eliez), University of Geneva School of Medicine, Geneva; Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva (Rochas, Seeber, Roehri, Rihs, Ferat, Michel); Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva (Schneider); Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland (Uhlhaas); Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin (Uhlhaas); Center for Biomedical Imaging (CIBM) of Lausanne and Geneva (Michel)
| |
Collapse
|
26
|
Karpinski BA, Maynard TM, Bryan CA, Yitsege G, Horvath A, Lee NH, Moody SA, LaMantia AS. Selective disruption of trigeminal sensory neurogenesis and differentiation in a mouse model of 22q11.2 deletion syndrome. Dis Model Mech 2022; 15:dmm047357. [PMID: 33722956 PMCID: PMC8126478 DOI: 10.1242/dmm.047357] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
22q11.2 Deletion Syndrome (22q11DS) is a neurodevelopmental disorder associated with cranial nerve anomalies and disordered oropharyngeal function, including pediatric dysphagia. Using the LgDel 22q11DS mouse model, we investigated whether sensory neuron differentiation in the trigeminal ganglion (CNgV), which is essential for normal orofacial function, is disrupted. We did not detect changes in cranial placode cell translocation or neural crest migration at early stages of LgDel CNgV development. However, as the ganglion coalesces, proportions of placode-derived LgDel CNgV cells increase relative to neural crest cells. In addition, local aggregation of placode-derived cells increases and aggregation of neural crest-derived cells decreases in LgDel CNgV. This change in cell-cell relationships was accompanied by altered proliferation of placode-derived cells at embryonic day (E)9.5, and premature neurogenesis from neural crest-derived precursors, reflected by an increased frequency of asymmetric neurogenic divisions for neural crest-derived precursors by E10.5. These early differences in LgDel CNgV genesis prefigure changes in sensory neuron differentiation and gene expression by postnatal day 8, when early signs of cranial nerve dysfunction associated with pediatric dysphagia are observed in LgDel mice. Apparently, 22q11 deletion destabilizes CNgV sensory neuron genesis and differentiation by increasing variability in cell-cell interaction, proliferation and sensory neuron differentiation. This early developmental divergence and its consequences may contribute to oropharyngeal dysfunction, including suckling, feeding and swallowing disruptions at birth, and additional orofacial sensory/motor deficits throughout life.
Collapse
Affiliation(s)
- Beverly A. Karpinski
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Thomas M. Maynard
- The Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA
| | - Corey A. Bryan
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Gelila Yitsege
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Anelia Horvath
- Department of Pharmacology and Physiology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Norman H. Lee
- Department of Pharmacology and Physiology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Sally A. Moody
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Anthony-Samuel LaMantia
- The Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
27
|
Tepper Á, Cuiza A, Alliende LM, Mena C, Ramirez-Mahaluf JP, Iruretagoyena B, Ornstein C, Fritsch R, Nachar R, González-Valderrama A, Undurraga J, Cruz JP, Tejos C, Fornito A, Repetto G, Crossley N. Functional Dysconnectivity in Ventral Striatocortical Systems in 22q11.2 Deletion Syndrome. Schizophr Bull 2021; 48:485-494. [PMID: 34931688 PMCID: PMC8886597 DOI: 10.1093/schbul/sbab139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
22q11.2 deletion syndrome (22q11.2DS) is a genetic neurodevelopmental disorder that represents one of the greatest known risk factors for psychosis. Previous studies in psychotic subjects without the deletion have identified a dopaminergic dysfunction in striatal regions, and dysconnectivity of striatocortical systems, as an important mechanism in the emergence of psychosis. Here, we used resting-state functional MRI to examine striatocortical functional connectivity in 22q11.2DS patients. We used a 2 × 2 factorial design including 125 subjects (55 healthy controls, 28 22q11.2DS patients without a history of psychosis, 10 22q11.2DS patients with a history of psychosis, and 32 subjects with a history of psychosis without the deletion), allowing us to identify network effects related to the deletion and to the presence of psychosis. In line with previous results from psychotic patients without 22q11.2DS, we found that there was a dorsal to ventral gradient of hypo- to hyperstriatocortical connectivity related to psychosis across both patient groups. The 22q11.2DS was additionally associated with abnormal functional connectivity in ventral striatocortical networks, with no significant differences identified in the dorsal system. Abnormalities in the ventral striatocortical system observed in these individuals with high genetic risk to psychosis may thus reflect a marker of illness risk.
Collapse
Affiliation(s)
- Ángeles Tepper
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Analía Cuiza
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luz María Alliende
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Mena
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile,Division of Psychology and Language Sciences, University College London, London, UK
| | | | - Barbara Iruretagoyena
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile,Department of Neurology and Psychiatry, Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Claudia Ornstein
- Hospital Clínico Universidad de Chile, Departamento de Psiquiatria y Salud Mental, Santiago, Chile
| | - Rosemarie Fritsch
- Hospital Clínico Universidad de Chile, Departamento de Psiquiatria y Salud Mental, Santiago, Chile
| | - Ruben Nachar
- Early Intervention Program, Instituto Psiquiátrico Dr J. Horwitz Barak, Santiago, Chile,School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Alfonso González-Valderrama
- Early Intervention Program, Instituto Psiquiátrico Dr J. Horwitz Barak, Santiago, Chile,School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Juan Undurraga
- Department of Neurology and Psychiatry, Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile,Early Intervention Program, Instituto Psiquiátrico Dr J. Horwitz Barak, Santiago, Chile
| | - Juan Pablo Cruz
- Department of Radiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Tejos
- Department of Electrical Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile,Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile,Biomedical Imaging Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Sciences and Monash Biomedical Imaging, Monash University, Melbourne, Australia
| | - Gabriela Repetto
- Genetic and Genomic Center, Universidad del Desarrollo, Santiago, Chile
| | - Nicolas Crossley
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile,Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile,Biomedical Imaging Center, Pontificia Universidad Católica de Chile, Santiago, Chile,To whom correspondence should be addressed; Diagonal Paraguay 362, Santiago, Chile; tel: 56 2 3543028, e-mail:
| |
Collapse
|
28
|
Räsänen N, Tiihonen J, Koskuvi M, Lehtonen Š, Koistinaho J. The iPSC perspective on schizophrenia. Trends Neurosci 2021; 45:8-26. [PMID: 34876311 DOI: 10.1016/j.tins.2021.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/29/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022]
Abstract
Over a decade of schizophrenia research using human induced pluripotent stem cell (iPSC)-derived neural models has provided substantial data describing neurobiological characteristics of the disorder in vitro. Simultaneously, translation of the results into general mechanistic concepts underlying schizophrenia pathophysiology has been trailing behind. Given that modeling brain function using cell cultures is challenging, the gap between the in vitro models and schizophrenia as a clinical disorder has remained wide. In this review, we highlight reproducible findings and emerging trends in recent schizophrenia-related iPSC studies. We illuminate the relevance of the results in the context of human brain development, with a focus on processes coinciding with critical developmental periods for schizophrenia.
Collapse
Affiliation(s)
- Noora Räsänen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Jari Tiihonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland; Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden; Center for Psychiatric Research, Stockholm City Council, Stockholm, Sweden; Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Marja Koskuvi
- Neuroscience Center, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, Helsinki, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
29
|
Lin A, Forsyth JK, Hoftman GD, Kushan-Wells L, Jalbrzikowski M, Dokuru D, Coppola G, Fiksinski A, Zinkstok J, Vorstman J, Nachun D, Bearden CE. Transcriptomic profiling of whole blood in 22q11.2 reciprocal copy number variants reveals that cell proportion highly impacts gene expression. Brain Behav Immun Health 2021; 18:100386. [PMID: 34841284 PMCID: PMC8607166 DOI: 10.1016/j.bbih.2021.100386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022] Open
Abstract
22q11.2 reciprocal copy number variants (CNVs) offer a powerful quasi-experimental "reverse-genetics" paradigm to elucidate how gene dosage (i.e., deletions and duplications) disrupts the transcriptome to cause further downstream effects. Clinical profiles of 22q11.2 CNV carriers indicate that disrupted gene expression causes alterations in neuroanatomy, cognitive function, and psychiatric disease risk. However, interpreting transcriptomic signal in bulk tissue requires careful consideration of potential changes in cell composition. We first characterized transcriptomic dysregulation in peripheral blood from reciprocal 22q11.2 CNV carriers using differential expression analysis and weighted gene co-expression network analysis (WGCNA) to identify modules of co-expressed genes. We also assessed for group differences in cell composition and re-characterized transcriptomic differences after accounting for cell type proportions and medication usage. Finally, to explore whether CNV-related transcriptomic changes relate to downstream phenotypes associated with 22q11.2 CNVs, we tested for associations of gene expression with neuroimaging measures and behavioral traits, including IQ and psychosis or ASD diagnosis. 22q11.2 deletion carriers (22qDel) showed widespread expression changes at the individual gene as well as module eigengene level compared to 22q11.2 duplication carriers (22qDup) and controls. 22qDup showed increased expression of 5 genes within the 22q11.2 locus, and CDH6 located outside of the locus. Downregulated modules in 22qDel implicated altered immune and inflammatory processes. Celltype deconvolution analyses revealed significant differences between CNV and control groups in T-cell, mast cell, and macrophage proportions; differential expression of individual genes between groups was substantially attenuated after adjusting for cell composition. Individual gene, module eigengene, and cell proportions were not significantly associated with psychiatric or neuroanatomic traits. Our findings suggest broad immune-related dysfunction in 22qDel and highlight the importance of understanding differences in cell composition when interpreting transcriptomic changes in clinical populations. Results also suggest novel directions for future investigation to test whether 22q11.2 CNV effects on macrophages have implications for brain-related microglial function that may contribute to psychiatric phenotypes in 22q11.2 CNV carriers.
Collapse
Affiliation(s)
- Amy Lin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jennifer K. Forsyth
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychology, University of Washington, WA, USA
| | - Gil D. Hoftman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leila Kushan-Wells
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Deepika Dokuru
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ania Fiksinski
- Wilhelmina Children's Hospital & University Medical Center Utrecht, Brain Center, the Netherlands
- Maastricht University, Department of Psychiatry and Neuropsychology, Division of Mental Health, MHeNS, the Netherlands
| | - Janneke Zinkstok
- Department of Psychiatry and Brain Center, University Medical Center Utrecht, the Netherlands
| | - Jacob Vorstman
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Daniel Nachun
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
30
|
Li Y, Xia Y, Zhu H, Luu E, Huang G, Sun Y, Sun K, Markx S, Leong KW, Xu B, Fu BM. Investigation of Neurodevelopmental Deficits of 22 q11.2 Deletion Syndrome with a Patient-iPSC-Derived Blood-Brain Barrier Model. Cells 2021; 10:cells10102576. [PMID: 34685556 PMCID: PMC8534009 DOI: 10.3390/cells10102576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/13/2022] Open
Abstract
The blood–brain barrier (BBB) is important in the normal functioning of the central nervous system. An altered BBB has been described in various neuropsychiatric disorders, including schizophrenia. However, the cellular and molecular mechanisms of such alterations remain unclear. Here, we investigate if BBB integrity is compromised in 22q11.2 deletion syndrome (also called DiGeorge syndrome), which is one of the validated genetic risk factors for schizophrenia. We utilized a set of human brain microvascular endothelial cells (HBMECs) derived from the induced pluripotent stem cell (iPSC) lines of patients with 22q11.2-deletion-syndrome-associated schizophrenia. We found that the solute permeability of the BBB formed from patient HBMECs increases by ~1.3–1.4-fold, while the trans-endothelial electrical resistance decreases to ~62% of the control values. Correspondingly, tight junction proteins and the endothelial glycocalyx that determine the integrity of the BBB are significantly disrupted. A transcriptome study also suggests that the transcriptional network related to the cell–cell junctions in the compromised BBB is substantially altered. An enrichment analysis further suggests that the genes within the altered gene expression network also contribute to neurodevelopmental disorders. Our findings suggest that neurovascular coupling can be targeted in developing novel therapeutical strategies for the treatment of 22q11.2 deletion syndrome.
Collapse
Affiliation(s)
- Yunfei Li
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA; (Y.L.); (Y.X.); (E.L.); (G.H.)
| | - Yifan Xia
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA; (Y.L.); (Y.X.); (E.L.); (G.H.)
| | - Huixiang Zhu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; (H.Z.); (Y.S.); (K.S.); (S.M.)
| | - Eric Luu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA; (Y.L.); (Y.X.); (E.L.); (G.H.)
| | - Guangyao Huang
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA; (Y.L.); (Y.X.); (E.L.); (G.H.)
| | - Yan Sun
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; (H.Z.); (Y.S.); (K.S.); (S.M.)
| | - Kevin Sun
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; (H.Z.); (Y.S.); (K.S.); (S.M.)
| | - Sander Markx
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; (H.Z.); (Y.S.); (K.S.); (S.M.)
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA;
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; (H.Z.); (Y.S.); (K.S.); (S.M.)
- Correspondence: (B.X.); (B.M.F.); Tel.: +1-212-650-7531 (B.M.F.)
| | - Bingmei M. Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA; (Y.L.); (Y.X.); (E.L.); (G.H.)
- Correspondence: (B.X.); (B.M.F.); Tel.: +1-212-650-7531 (B.M.F.)
| |
Collapse
|
31
|
Favicchia I, Flore G, Cioffi S, Lania G, Baldini A, Illingworth E. Pharmacological Rescue of the Brain Cortex Phenotype of Tbx1 Mouse Mutants: Significance for 22q11.2 Deletion Syndrome. Front Mol Neurosci 2021; 14:663598. [PMID: 34552467 PMCID: PMC8450345 DOI: 10.3389/fnmol.2021.663598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives Tbx1 mutant mice are a widely used model of 22q11.2 deletion syndrome (22q11.2DS) because they manifest a broad spectrum of physical and behavioral abnormalities that is similar to that found in 22q11.2DS patients. In Tbx1 mutants, brain abnormalities include changes in cortical cytoarchitecture, hypothesized to be caused by the precocious differentiation of cortical progenitors. The objectives of this research are to identify drugs that have efficacy against the brain phenotype, and through a phenotypic rescue approach, gain insights into the pathogenetic mechanisms underlying Tbx1 haploinsufficiency. Experimental Approach Disease model: Tbx1 heterozygous and homozygous embryos. We tested the ability of two FDA-approved drugs, the LSD1 inhibitor Tranylcypromine and Vitamin B12, to rescue the Tbx1 mutant cortical phenotype. Both drugs have proven efficacy against the cardiovascular phenotype, albeit at a much reduced level compared to the rescue achieved in the brain. Methods In situ hybridization and immunostaining of histological brain sections using a subset of molecular markers that label specific cortical regions or cell types. Appropriate quantification and statistical analysis of gene and protein expression were applied to identify cortical abnormalities and to determine the level of phenotypic rescue achieved. Results Cortical abnormalities observed in Tbx1 mutant embryos were fully rescued by both drugs. Intriguingly, rescue was obtained with both drugs in Tbx1 homozygous mutants, indicating that they function through mechanisms that do not depend upon Tbx1 function. This was particularly surprising for Vitamin B12, which was identified through its ability to increase Tbx1 gene expression. Conclusion To our knowledge, this is only the second example of drugs to be identified that ameliorate phenotypes caused by the mutation of a single gene from the 22q11.2 homologous region of the mouse genome. This one drug-one gene approach might be important because there is evidence that the brain phenotype in 22q11.2DS patients is multigenic in origin, unlike the physical phenotypes, which are overwhelmingly attributable to Tbx1 haploinsufficiency. Therefore, effective treatments will likely involve the use of multiple drugs that are targeted to the function of specific genes within the deleted region.
Collapse
Affiliation(s)
- Ilaria Favicchia
- Department of Chemistry and Biology, "Adolfo Zambelli", University of Salerno, Fisciano, Italy
| | - Gemma Flore
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Naples, Italy
| | - Sara Cioffi
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Naples, Italy
| | - Gabriella Lania
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Naples, Italy
| | - Antonio Baldini
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples, Federico II, Naples, Italy
| | - Elizabeth Illingworth
- Department of Chemistry and Biology, "Adolfo Zambelli", University of Salerno, Fisciano, Italy
| |
Collapse
|
32
|
Motahari Z, Maynard TM, Popratiloff A, Moody SA, LaMantia AS. Aberrant early growth of individual trigeminal sensory and motor axons in a series of mouse genetic models of 22q11.2 deletion syndrome. Hum Mol Genet 2021; 29:3081-3093. [PMID: 32901287 PMCID: PMC7645708 DOI: 10.1093/hmg/ddaa199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/29/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
We identified divergent modes of initial axon growth that prefigure disrupted differentiation of the trigeminal nerve (CN V), a cranial nerve essential for suckling, feeding and swallowing (S/F/S), a key innate behavior compromised in multiple genetic developmental disorders including DiGeorge/22q11.2 Deletion Syndrome (22q11.2 DS). We combined rapid in vivo labeling of single CN V axons in LgDel+/− mouse embryos, a genomically accurate 22q11.2DS model, and 3D imaging to identify and quantify phenotypes that could not be resolved using existing methods. We assessed these phenotypes in three 22q11.2-related genotypes to determine whether individual CN V motor and sensory axons wander, branch and sprout aberrantly in register with altered anterior–posterior hindbrain patterning and gross morphological disruption of CN V seen in LgDel+/−. In the additional 22q11.2-related genotypes: Tbx1+/−, Ranbp1−/−, Ranbp1+/− and LgDel+/−:Raldh2+/−; axon phenotypes are seen when hindbrain patterning and CN V gross morphology is altered, but not when it is normal or restored toward WT. This disordered growth of CN V sensory and motor axons, whose appropriate targeting is critical for optimal S/F/S, may be an early, critical determinant of imprecise innervation leading to inefficient oropharyngeal function associated with 22q11.2 deletion from birth onward.
Collapse
Affiliation(s)
- Zahra Motahari
- Institute for Neuroscience, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Thomas M Maynard
- The Fralin Biomedical Research Institute at Virginia Tech-Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Anastas Popratiloff
- Institute for Neuroscience, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Sally A Moody
- Institute for Neuroscience, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Anthony-S LaMantia
- The Fralin Biomedical Research Institute at Virginia Tech-Carilion School of Medicine, Roanoke, VA 24016, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
33
|
Moreau CA, Raznahan A, Bellec P, Chakravarty M, Thompson PM, Jacquemont S. Dissecting autism and schizophrenia through neuroimaging genomics. Brain 2021; 144:1943-1957. [PMID: 33704401 PMCID: PMC8370419 DOI: 10.1093/brain/awab096] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
Neuroimaging genomic studies of autism spectrum disorder and schizophrenia have mainly adopted a 'top-down' approach, beginning with the behavioural diagnosis, and moving down to intermediate brain phenotypes and underlying genetic factors. Advances in imaging and genomics have been successfully applied to increasingly large case-control studies. As opposed to diagnostic-first approaches, the bottom-up strategy begins at the level of molecular factors enabling the study of mechanisms related to biological risk, irrespective of diagnoses or clinical manifestations. The latter strategy has emerged from questions raised by top-down studies: why are mutations and brain phenotypes over-represented in individuals with a psychiatric diagnosis? Are they related to core symptoms of the disease or to comorbidities? Why are mutations and brain phenotypes associated with several psychiatric diagnoses? Do they impact a single dimension contributing to all diagnoses? In this review, we aimed at summarizing imaging genomic findings in autism and schizophrenia as well as neuropsychiatric variants associated with these conditions. Top-down studies of autism and schizophrenia identified patterns of neuroimaging alterations with small effect-sizes and an extreme polygenic architecture. Genomic variants and neuroimaging patterns are shared across diagnostic categories suggesting pleiotropic mechanisms at the molecular and brain network levels. Although the field is gaining traction; characterizing increasingly reproducible results, it is unlikely that top-down approaches alone will be able to disentangle mechanisms involved in autism or schizophrenia. In stark contrast with top-down approaches, bottom-up studies showed that the effect-sizes of high-risk neuropsychiatric mutations are equally large for neuroimaging and behavioural traits. Low specificity has been perplexing with studies showing that broad classes of genomic variants affect a similar range of behavioural and cognitive dimensions, which may be consistent with the highly polygenic architecture of psychiatric conditions. The surprisingly discordant effect sizes observed between genetic and diagnostic first approaches underscore the necessity to decompose the heterogeneity hindering case-control studies in idiopathic conditions. We propose a systematic investigation across a broad spectrum of neuropsychiatric variants to identify putative latent dimensions underlying idiopathic conditions. Gene expression data on temporal, spatial and cell type organization in the brain have also considerable potential for parsing the mechanisms contributing to these dimensions' phenotypes. While large neuroimaging genomic datasets are now available in unselected populations, there is an urgent need for data on individuals with a range of psychiatric symptoms and high-risk genomic variants. Such efforts together with more standardized methods will improve mechanistically informed predictive modelling for diagnosis and clinical outcomes.
Collapse
Affiliation(s)
- Clara A Moreau
- Sainte Justine Research Center, University of Montréal, Montréal, Québec H3T 1C5, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, Québec H3W 1W5, Canada
- Human Genetics and Cognitive Functions, CNRS UMR 3571, Université de Paris, Institut Pasteur, Paris, France
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, MD 20892, USA
| | - Pierre Bellec
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, Québec H3W 1W5, Canada
| | - Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Hospital Mental Health University Institute, Verdun, Québec H4H 1R3, Canada
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Marina del Rey, CA 90033, USA
| | - Sebastien Jacquemont
- Sainte Justine Research Center, University of Montréal, Montréal, Québec H3T 1C5, Canada
| |
Collapse
|
34
|
Maynard TM, Horvath A, Bernot JP, Karpinski BA, Tavares ALP, Shah A, Zheng Q, Spurr L, Olender J, Moody SA, Fraser CM, LaMantia AS, Lee NH. Transcriptional dysregulation in developing trigeminal sensory neurons in the LgDel mouse model of DiGeorge 22q11.2 deletion syndrome. Hum Mol Genet 2021; 29:1002-1017. [PMID: 32047912 PMCID: PMC7158380 DOI: 10.1093/hmg/ddaa024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/12/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
LgDel mice, which model the heterozygous deletion of genes at human chromosome 22q11.2 associated with DiGeorge/22q11.2 deletion syndrome (22q11DS), have cranial nerve and craniofacial dysfunction as well as disrupted suckling, feeding and swallowing, similar to key 22q11DS phenotypes. Divergent trigeminal nerve (CN V) differentiation and altered trigeminal ganglion (CNgV) cellular composition prefigure these disruptions in LgDel embryos. We therefore asked whether a distinct transcriptional state in a specific population of early differentiating LgDel cranial sensory neurons, those in CNgV, a major source of innervation for appropriate oropharyngeal function, underlies this departure from typical development. LgDel versus wild-type (WT) CNgV transcriptomes differ significantly at E10.5 just after the ganglion has coalesced. Some changes parallel altered proportions of cranial placode versus cranial neural crest-derived CNgV cells. Others are consistent with a shift in anterior-posterior patterning associated with divergent LgDel cranial nerve differentiation. The most robust quantitative distinction, however, is statistically verifiable increased variability of expression levels for most of the over 17 000 genes expressed in common in LgDel versus WT CNgV. Thus, quantitative expression changes of functionally relevant genes and increased stochastic variation across the entire CNgV transcriptome at the onset of CN V differentiation prefigure subsequent disruption of cranial nerve differentiation and oropharyngeal function in LgDel mice.
Collapse
Affiliation(s)
- Thomas M Maynard
- Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, 24016 USA.,Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Anelia Horvath
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.,McCormick Genomics and Proteomics Center, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - James P Bernot
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Beverly A Karpinski
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Andre L P Tavares
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Ankita Shah
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Qianqian Zheng
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Liam Spurr
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Jacqueline Olender
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Sally A Moody
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Claire M Fraser
- Institute for Genome Sciences, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Anthony-S LaMantia
- Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, 24016 USA.,Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.,Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg VA, 24061, USA.,Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Norman H Lee
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
35
|
Lin I, Afshar Y, Goldstein J, Grossman J, Grody WW, Quintero-Rivera F. Central 22q11.2 deletion (LCR22 B-D) in a fetus with severe fetal growth restriction and a mother with severe systemic lupus erythematosus: Further evidence of CRKL haploinsufficiency in the pathogenesis of 22q11.2 deletion syndrome. Am J Med Genet A 2021; 185:3042-3047. [PMID: 34196458 DOI: 10.1002/ajmg.a.62346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/03/2021] [Accepted: 04/22/2021] [Indexed: 12/25/2022]
Abstract
22q11.2 deletion syndrome (22q11.2 DS, MIM #188400) is the most common chromosomal microdeletion with an incidence of 1 in 4000 live births. 22q11.2 DS patients present with varying penetrance and a broad phenotypic spectrum including dysmorphic features, congenital heart defects, hypoplastic thymus and T-cell deficiency, and hypocalcemia. The typical deletion spans 3 Mb between 4 large blocks of repetitive DNA, known as low copy repeats (LCRs), on chromosome 22 (LCR22) A and D. This deletion is found in ~85% of 22q11.2 DS patients, while only 4-5% have central LCR22B-D (1.5 Mb) and LCR22C-D (0.7 Mb) deletions. We report on a prenatally diagnosed, inherited case of central LCR22B-D 22q11.2 DS, born to a 22-year-old female with multiple autoimmune disorders. These include Sjogren's-syndrome-related antigen A (SSA+) severe systemic lupus erythematosus (SLE) with cutaneous and discoid components and seronegative antiphospholipid syndrome. Amniocentesis was performed due to fetal growth restriction (FGR). FISH with TUPLE1 (HIRA) probe was normal; however, chromosomal microarray identified a ~737 kb heterozygous loss between LCR22B-D. Subsequently, the same deletion was identified in the mother, which included CRKL and 19 other genes but excluded HIRA and TBX1, the typical candidate genes for 22q11.2DS pathogenesis. This case explores how loss of CRKL may contribute to immune dysregulation, as seen in the multiple severe autoimmune phenotypes of the mother, and FGR. Our experience confirms the importance of thorough workup in individuals with reduced penetrance of 22q11.2 DS features or atypical clinical presentations.
Collapse
Affiliation(s)
- Isabella Lin
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Yalda Afshar
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of California Los Angeles, Los Angeles, California, USA
| | - Jeffrey Goldstein
- Pathology and Laboratory Medicine, UCLA Center for Health Sciences, Los Angeles, California, USA
| | - Jennifer Grossman
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Division of Rheumatology, Department of Medicine, University of California Los Angeles, California, USA
| | - Wayne W Grody
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Pathology and Laboratory Medicine, UCLA Center for Health Sciences, Los Angeles, California, USA.,Department of Human Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Fabiola Quintero-Rivera
- Departments of Pathology and Laboratory Medicine and Pediatrics, Division of Genetic and Genomic Medicine, University of California, Irvine, California, USA
| |
Collapse
|
36
|
Total Anomalous Pulmonary Venous Connection in Mother and Son with a Central 22q11.2 Microdeletion. Case Rep Genet 2021; 2021:5539855. [PMID: 34221520 PMCID: PMC8213480 DOI: 10.1155/2021/5539855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/14/2021] [Accepted: 06/04/2021] [Indexed: 11/17/2022] Open
Abstract
In this clinical report, we describe a male infant and his mother, who had similar congenital heart defects. They were both diagnosed neonatally with total anomalous pulmonary venous connection (TAPVC) in combination with other heart defects. Neither of the two had any other organ malformations or dysmorphic facial features. SNP-array identified a central 22q11.2 microdeletion in the male infant and his mother as well as in the maternal grandmother and maternal aunt. The mother and the maternal aunt additionally harbored a 15q11.2 BP1-BP2 microdeletion. The maternal grandmother was unaffected by heart disease. However, heart computed tomography scan of the maternal aunt revealed a quadricuspid aortic valve. Additionally, the maternal grandmother and the maternal aunt both had significant learning disabilities. Rarely, TAPVC has been described in patients with the common 22q11.2 microdeletions. However, to the best of our knowledge, TAPVC has not previously been reported in patients with this small central 22q11.2 microdeletion. Haploinsufficiency of TBX1 was originally thought to be the main cause of the 22q11.2 microdeletion syndrome phenotype, but TBX1 is not included in the atypical central 22q11.2 microdeletion. Previous reports have suggested an association between TAPVC and the 15q11.2 BP1-BP2 microdeletion. Our report does not support this association as the maternal aunt, who harbors both microdeletions, is unaffected by TAPVC, and the male infant affected by TAPVC does not harbor the 15q11.2 BP1-BP2 microdeletion. Our findings support that genes located in the central 22q11.2 region are important for heart development and that haploinsufficiency of these genes plays a crucial role in the development of the rare heart defect TAPVC.
Collapse
|
37
|
Forsyth JK, Mennigen E, Lin A, Sun D, Vajdi A, Kushan-Wells L, Ching CRK, Villalon-Reina JE, Thompson PM, Bearden CE. Prioritizing Genetic Contributors to Cortical Alterations in 22q11.2 Deletion Syndrome Using Imaging Transcriptomics. Cereb Cortex 2021; 31:3285-3298. [PMID: 33638978 PMCID: PMC8196250 DOI: 10.1093/cercor/bhab008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/13/2020] [Accepted: 05/02/2020] [Indexed: 11/25/2022] Open
Abstract
22q11.2 deletion syndrome (22q11DS) results from a hemizygous deletion that typically spans 46 protein-coding genes and is associated with widespread alterations in brain morphology. The specific genetic mechanisms underlying these alterations remain unclear. In the 22q11.2 ENIGMA Working Group, we characterized cortical alterations in individuals with 22q11DS (n = 232) versus healthy individuals (n = 290) and conducted spatial convergence analyses using gene expression data from the Allen Human Brain Atlas to prioritize individual genes that may contribute to altered surface area (SA) and cortical thickness (CT) in 22q11DS. Total SA was reduced in 22q11DS (Z-score deviance = -1.04), with prominent reductions in midline posterior and lateral association regions. Mean CT was thicker in 22q11DS (Z-score deviance = +0.64), with focal thinning in a subset of regions. Regional expression of DGCR8 was robustly associated with regional severity of SA deviance in 22q11DS; AIFM3 was also associated with SA deviance. Conversely, P2RX6 was associated with CT deviance. Exploratory analysis of gene targets of microRNAs previously identified as down-regulated due to DGCR8 deficiency suggested that DGCR8 haploinsufficiency may contribute to altered corticogenesis in 22q11DS by disrupting cell cycle modulation. These findings demonstrate the utility of combining neuroanatomic and transcriptomic datasets to derive molecular insights into complex, multigene copy number variants.
Collapse
Affiliation(s)
- Jennifer K Forsyth
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
| | - Eva Mennigen
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Amy Lin
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
- Interdepartmental Neuroscience Program, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Daqiang Sun
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Ariana Vajdi
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
| | - Leila Kushan-Wells
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Julio E Villalon-Reina
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Carrie E Bearden
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA 90024, USA
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
38
|
Fiksinski AM, Schneider M, Zinkstok J, Baribeau D, Chawner SJRA, Vorstman JAS. Neurodevelopmental Trajectories and Psychiatric Morbidity: Lessons Learned From the 22q11.2 Deletion Syndrome. Curr Psychiatry Rep 2021; 23:13. [PMID: 33625600 PMCID: PMC7904715 DOI: 10.1007/s11920-021-01225-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The 22q11.2 deletion syndrome (22q11DS) is associated with a broad spectrum of neurodevelopmental phenotypes and is the strongest known single genetic risk factor for schizophrenia. Compared to other rare structural pathogenic genetic variants, 22q11DS is relatively common and one of the most extensively studied. This review provides a state-of-the-art overview of current insights regarding associated neurodevelopmental phenotypes and potential implications for 22q11DS and beyond. RECENT FINDINGS We will first discuss recent findings with respect to neurodevelopmental phenotypic expression associated with 22q11DS, including psychotic disorders, intellectual functioning, autism spectrum disorders, as well as their interactions. Second, we will address considerations that are important in interpreting these data and propose potential implications for both the clinical care for and the empirical study of individuals with 22q11DS. Third, we will highlight variable penetrance and pleiotropy with respect to neurodevelopmental phenotypes in 22q11DS. We will discuss how these phenomena are consistently observed in the context of virtually all rare pathogenic variants and that they pose substantial challenges from both a clinical and a research perspective. We outline how 22q11DS could be viewed as a genetic model for studying neurodevelopmental phenotypes. In addition, we propose that 22q11DS research can help elucidate mechanisms underlying variable expression and pleiotropy of neurodevelopmental phenotypes, insights that are likely relevant for 22q11DS and beyond, including for individuals with other rare pathogenic genetic variants and for individuals with idiopathic neurodevelopmental conditions.
Collapse
Affiliation(s)
- Ania M. Fiksinski
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
- Dalglish Family 22q Clinic for Adults with 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, Canada
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario Canada
| | - Maude Schneider
- Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva, Switzerland
- Department of Neurosciences, Center for Contextual Psychiatry, KU Leuven, Leuven, Belgium
| | - Janneke Zinkstok
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Danielle Baribeau
- Department of Psychiatry, Hospital for Sick Children, Toronto, ON Canada
- Department of Psychiatry, University of Toronto, Toronto, ON Canada
| | - Samuel J. R. A. Chawner
- Cardiff University Centre for Human Developmental Science, School of Psychology, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Jacob A. S. Vorstman
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, Hospital for Sick Children, Toronto, ON Canada
- Department of Psychiatry, University of Toronto, Toronto, ON Canada
- The Centre for Applied Genomics, Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada
| |
Collapse
|
39
|
Peter B, Scherer N, Liang WS, Pophal S, Nielsen C, Grebe TA. A phenotypically diverse family with an atypical 22q11.2 deletion due to an unbalanced 18q23;22q11.2 translocation. Am J Med Genet A 2021; 185:1532-1537. [PMID: 33569883 DOI: 10.1002/ajmg.a.62121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/03/2021] [Accepted: 01/24/2021] [Indexed: 11/08/2022]
Abstract
The 22q11.2 deletion syndrome (22q11.2 DS) is the most common deletion syndrome in humans. In most cases, it occurs de novo. A rare family of three with 22q11.2 deletion syndrome (22q11.2 DS) resulting from an unbalanced 18q;22q translocation is reported here. Their deletion region is atypical in that it includes only 26 of the 36 genes in the minimal critical 22q11.2 DS region but it involves the loss of the centromeric 22q region and the entire p arm. The deletion region overlaps with seven other rare atypical cases; common to all cases was the loss of a region including SEPT5-GP1BB proximally and most of ARVCF distally. Interrogation of the deleted 22q region proximal to the canonical 22q11.2 deletion region in the DECIPHER database showed seven cases with isolated or combined traits of 22q11.2 DS, including three with clefts. The phenotypes in the present family thus may result from the loss of a subset of genes in the critical region, or alternatively the loss of other genes or sequences in the proximal 22q deletion region, or interactive effects among these. Despite the identical deletion locus in the three affected family members, expression of the 22q11.2 DS traits differed substantially among them. These three related cases thus contribute to knowledge of 22q11.2 DS in that their unusual deletion locus co-occurred with the cardinal features of the syndrome while their identical deletions are associated with variable phenotypic expression.
Collapse
Affiliation(s)
- Beate Peter
- Speech and Hearing Science, College of Health Solutions, Arizona State University, Tempe, Arizona, USA.,Department of Communication Sciences and Disorders, Saint Louis University, Saint Louis, Missouri, USA
| | - Nancy Scherer
- Speech and Hearing Science, College of Health Solutions, Arizona State University, Tempe, Arizona, USA
| | - Winnie S Liang
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Stephen Pophal
- Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Colby Nielsen
- College of Medicine, University of Arizona, Phoenix, Arizona, USA
| | - Theresa A Grebe
- Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Arizona, USA
| |
Collapse
|
40
|
Manno GC, Segal GS, Yu A, Xu F, Ray JW, Cooney E, Britt AD, Jain SK, Goldblum RM, Robinson SS, Dong J. Genotypic and phenotypic variability of 22q11.2 microdeletions – an institutional experience. AIMS MOLECULAR SCIENCE 2021; 8:257-274. [PMID: 34938854 PMCID: PMC8691803 DOI: 10.3934/molsci.2021020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Patients with chromosome 22q11.2 deletion syndromes classically present with variable cardiac defects, parathyroid and thyroid gland hypoplasia, immunodeficiency and velopharyngeal insufficiency, developmental delay, intellectual disability, cognitive impairment, and psychiatric disorders. New technologies including chromosome microarray have identified smaller deletions in the 22q11.2 region. An increasing number of studies have reported patients presenting with various features harboring smaller 22q11.2 deletions, suggesting a need to better elucidate 22q11.2 deletions and their phenotypic contributions so that clinicians may better guide prognosis for families. We identified 16 pediatric patients at our institution harboring various 22q11.2 deletions detected by chromosomal microarray and report their clinical presentations. Findings include various neurodevelopmental delays with the most common one being attention deficit hyperactivity disorder (ADHD), one reported case of infant lethality, four cases of preterm birth, one case with dual diagnoses of 22q11.2 microdeletion and Down syndrome. We examined potential genotypic contributions of the deleted regions.</p>
</abstract>
Collapse
Affiliation(s)
- Gabrielle C. Manno
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Gabrielle S. Segal
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alexander Yu
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Fangling Xu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Joseph W. Ray
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Erin Cooney
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Allison D. Britt
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sunil K. Jain
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Randall M. Goldblum
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sally S. Robinson
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jianli Dong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Correspondence:; Tel: 4097724866
| |
Collapse
|
41
|
LaMantia AS. Why Does the Face Predict the Brain? Neural Crest Induction, Craniofacial Morphogenesis, and Neural Circuit Development. Front Physiol 2020; 11:610970. [PMID: 33362582 PMCID: PMC7759552 DOI: 10.3389/fphys.2020.610970] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchephalic and rhombencephalic neural crest cells generate the craniofacial skeleton, special sensory organs, and subsets of cranial sensory receptor neurons. They do so while preserving the anterior-posterior (A-P) identity of their neural tube origins. This organizational principle is paralleled by central nervous system circuits that receive and process information from facial structures whose A-P identity is in register with that in the brain. Prior to morphogenesis of the face and its circuits, however, neural crest cells act as "inductive ambassadors" from distinct regions of the neural tube to induce differentiation of target craniofacial domains and establish an initial interface between the brain and face. At every site of bilateral, non-axial secondary induction, neural crest constitutes all or some of the mesenchymal compartment for non-axial mesenchymal/epithelial (M/E) interactions. Thus, for epithelial domains in the craniofacial primordia, aortic arches, limbs, the spinal cord, and the forebrain (Fb), neural crest-derived mesenchymal cells establish local sources of inductive signaling molecules that drive morphogenesis and cellular differentiation. This common mechanism for building brains, faces, limbs, and hearts, A-P axis specified, neural crest-mediated M/E induction, coordinates differentiation of distal structures, peripheral neurons that provide their sensory or autonomic innervation in some cases, and central neural circuits that regulate their behavioral functions. The essential role of this neural crest-mediated mechanism identifies it as a prime target for pathogenesis in a broad range of neurodevelopmental disorders. Thus, the face and the brain "predict" one another, and this mutual developmental relationship provides a key target for disruption by developmental pathology.
Collapse
Affiliation(s)
- Anthony-Samuel LaMantia
- Laboratory of Developmental Disorders and Genetics and Center for Neurobiology Research, Fralin Biomedical Research Institute, Department of Pediatrics, Virginia Tech-Carilion School of Medicine, Virginia Tech, Roanoke, VA, United States.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
42
|
Cirillo E, Prencipe MR, Giardino G, Romano R, Scalia G, Genesio R, Nitsch L, Pignata C. Clinical Phenotype, Immunological Abnormalities, and Genomic Findings in Patients with DiGeorge Spectrum Phenotype without 22q11.2 Deletion. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 8:3112-3120. [DOI: 10.1016/j.jaip.2020.06.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023]
|
43
|
Yitsege G, Stokes BA, Sabatino JA, Sugrue KF, Banyai G, Paronett EM, Karpinski BA, Maynard TM, LaMantia A, Zohn IE. Variations in maternal vitamin A intake modifies phenotypes in a mouse model of 22q11.2 deletion syndrome. Birth Defects Res 2020; 112:1194-1208. [PMID: 32431076 PMCID: PMC7586978 DOI: 10.1002/bdr2.1709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/17/2020] [Accepted: 04/25/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vitamin A regulates patterning of the pharyngeal arches, cranial nerves, and hindbrain that are essential for feeding and swallowing. In the LgDel mouse model of 22q11.2 deletion syndrome (22q11DS), morphogenesis of multiple structures involved in feeding and swallowing are dysmorphic. We asked whether changes in maternal dietary Vitamin A intake can modify cranial nerve, hindbrain and pharyngeal arch artery development in the embryo as well as lung pathology that can be a sign of aspiration dysphagia in LgDel pups. METHODS Three defined amounts of vitamin A (4, 10, and 16 IU/g) were provided in the maternal diet. Cranial nerve, hindbrain and pharyngeal arch artery development was evaluated in embryos and inflammation in the lungs of pups to determine the impact of altering maternal diet on these phenotypes. RESULTS Reduced maternal vitamin A intake improved whereas increased intake exacerbated lung inflammation in LgDel pups. These changes were accompanied by increased incidence and/or severity of pharyngeal arch artery and cranial nerve V (CN V) abnormalities in LgDel embryos as well as altered expression of Cyp26b1 in the hindbrain. CONCLUSIONS Our studies demonstrate that variations in maternal vitamin A intake can influence the incidence and severity of phenotypes in a mouse model 22q11.2 deletion syndrome.
Collapse
Affiliation(s)
- Gelila Yitsege
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Center for Genetic MedicineChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Bethany A. Stokes
- Center for Neuroscience ResearchChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
- Center for Genetic MedicineChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Julia A. Sabatino
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Kelsey F. Sugrue
- Center for Neuroscience ResearchChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
- Center for Genetic MedicineChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Gabor Banyai
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Center for Neuroscience ResearchChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Elizabeth M. Paronett
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Beverly A. Karpinski
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Thomas M. Maynard
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of MedicineRoanokeVirginiaUSA
| | - Anthony‐S. LaMantia
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of MedicineRoanokeVirginiaUSA
- Department of Biological SciencesVirginia TechBlacksburgVirginiaUSA
| | - Irene E. Zohn
- Institute for NeuroscienceThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
- Center for Neuroscience ResearchChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
- Center for Genetic MedicineChildren’s Research Institute, Children’s National Medical CenterWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
44
|
Comer AL, Carrier M, Tremblay MÈ, Cruz-Martín A. The Inflamed Brain in Schizophrenia: The Convergence of Genetic and Environmental Risk Factors That Lead to Uncontrolled Neuroinflammation. Front Cell Neurosci 2020; 14:274. [PMID: 33061891 PMCID: PMC7518314 DOI: 10.3389/fncel.2020.00274] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a disorder with a heterogeneous etiology involving complex interplay between genetic and environmental risk factors. The immune system is now known to play vital roles in nervous system function and pathology through regulating neuronal and glial development, synaptic plasticity, and behavior. In this regard, the immune system is positioned as a common link between the seemingly diverse genetic and environmental risk factors for schizophrenia. Synthesizing information about how the immune-brain axis is affected by multiple factors and how these factors might interact in schizophrenia is necessary to better understand the pathogenesis of this disease. Such knowledge will aid in the development of more translatable animal models that may lead to effective therapeutic interventions. Here, we provide an overview of the genetic risk factors for schizophrenia that modulate immune function. We also explore environmental factors for schizophrenia including exposure to pollution, gut dysbiosis, maternal immune activation and early-life stress, and how the consequences of these risk factors are linked to microglial function and dysfunction. We also propose that morphological and signaling deficits of the blood-brain barrier, as observed in some individuals with schizophrenia, can act as a gateway between peripheral and central nervous system inflammation, thus affecting microglia in their essential functions. Finally, we describe the diverse roles that microglia play in response to neuroinflammation and their impact on brain development and homeostasis, as well as schizophrenia pathophysiology.
Collapse
Affiliation(s)
- Ashley L. Comer
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
- Neurophotonics Center, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Alberto Cruz-Martín
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
- Neurophotonics Center, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States
| |
Collapse
|
45
|
Consequences of 22q11.2 Microdeletion on the Genome, Individual and Population Levels. Genes (Basel) 2020; 11:genes11090977. [PMID: 32842603 PMCID: PMC7563277 DOI: 10.3390/genes11090977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/27/2022] Open
Abstract
Chromosomal 22q11.2 deletion syndrome (22q11.2DS) (ORPHA: 567) caused by microdeletion in chromosome 22 is the most common chromosomal microdeletion disorder in humans. Despite the same change on the genome level, like in the case of monozygotic twins, phenotypes are expressed differently in 22q11.2 deletion individuals. The rest of the genome, as well as epigenome and environmental factors, are not without influence on the variability of phenotypes. The penetrance seems to be more genotype specific than deleted locus specific. The transcript levels of deleted genes are not usually reduced by 50% as assumed due to haploinsufficiency. 22q11.2DS is often an undiagnosed condition, as each patient may have a different set out of 180 possible clinical manifestations. Diverse dysmorphic traits are present in patients from different ethnicities, which makes diagnosis even more difficult. 22q11.2 deletion syndrome serves as an example of a genetic syndrome that is not easy to manage at all stages: diagnosis, consulting and dealing with.
Collapse
|
46
|
Cardiac Neural Crest Cells: Their Rhombomeric Specification, Migration, and Association with Heart and Great Vessel Anomalies. Cell Mol Neurobiol 2020; 41:403-429. [PMID: 32405705 DOI: 10.1007/s10571-020-00863-w] [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: 10/07/2019] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Outflow tract abnormalities are the most frequent congenital heart defects. These are due to the absence or dysfunction of the two main cell types, i.e., neural crest cells and secondary heart field cells that migrate in opposite directions at the same stage of development. These cells directly govern aortic arch patterning and development, ascending aorta dilatation, semi-valvular and coronary artery development, aortopulmonary septation abnormalities, persistence of the ductus arteriosus, trunk and proximal pulmonary arteries, sub-valvular conal ventricular septal/rotational defects, and non-compaction of the left ventricle. In some cases, depending on the functional defects of these cells, additional malformations are found in the expected spatial migratory area of the cells, namely in the pharyngeal arch derivatives and cervico-facial structures. Associated non-cardiovascular anomalies are often underestimated, since the multipotency and functional alteration of these cells can result in the modification of multiple neural, epidermal, and cervical structures at different levels. In most cases, patients do not display the full phenotype of abnormalities, but congenital cardiac defects involving the ventricular outflow tract, ascending aorta, aortic arch and supra-aortic trunks should be considered as markers for possible impaired function of these cells. Neural crest cells should not be considered as a unique cell population but on the basis of their cervical rhombomere origins R3-R5 or R6-R7-R8 and specific migration patterns: R3-R4 towards arch II, R5-R6 arch III and R7-R8 arch IV and VI. A better understanding of their development may lead to the discovery of unknown associated abnormalities, thereby enabling potential improvements to be made to the therapeutic approach.
Collapse
|
47
|
Maynard TM, Zohn IE, Moody SA, LaMantia AS. Suckling, Feeding, and Swallowing: Behaviors, Circuits, and Targets for Neurodevelopmental Pathology. Annu Rev Neurosci 2020; 43:315-336. [PMID: 32101484 DOI: 10.1146/annurev-neuro-100419-100636] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
All mammals must suckle and swallow at birth, and subsequently chew and swallow solid foods, for optimal growth and health. These initially innate behaviors depend critically upon coordinated development of the mouth, tongue, pharynx, and larynx as well as the cranial nerves that control these structures. Disrupted suckling, feeding, and swallowing from birth onward-perinatal dysphagia-is often associated with several neurodevelopmental disorders that subsequently alter complex behaviors. Apparently, a broad range of neurodevelopmental pathologic mechanisms also target oropharyngeal and cranial nerve differentiation. These aberrant mechanisms, including altered patterning, progenitor specification, and neurite growth, prefigure dysphagia and may then compromise circuits for additional behavioral capacities. Thus, perinatal dysphagia may be an early indicator of disrupted genetic and developmental programs that compromise neural circuits and yield a broad range of behavioral deficits in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Thomas M Maynard
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016, USA;
| | - Irene E Zohn
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.,Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20037, USA
| | - Sally A Moody
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Anthony-S LaMantia
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016, USA; .,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia 24061, USA
| |
Collapse
|
48
|
Eom TY, Han SB, Kim J, Blundon JA, Wang YD, Yu J, Anderson K, Kaminski DB, Sakurada SM, Pruett-Miller SM, Horner L, Wagner B, Robinson CG, Eicholtz M, Rose DC, Zakharenko SS. Schizophrenia-related microdeletion causes defective ciliary motility and brain ventricle enlargement via microRNA-dependent mechanisms in mice. Nat Commun 2020; 11:912. [PMID: 32060266 PMCID: PMC7021727 DOI: 10.1038/s41467-020-14628-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/22/2020] [Indexed: 01/11/2023] Open
Abstract
Progressive ventricular enlargement, a key feature of several neurologic and psychiatric diseases, is mediated by unknown mechanisms. Here, using murine models of 22q11-deletion syndrome (22q11DS), which is associated with schizophrenia in humans, we found progressive enlargement of lateral and third ventricles and deceleration of ciliary beating on ependymal cells lining the ventricular walls. The cilia-beating deficit observed in brain slices and in vivo is caused by elevated levels of dopamine receptors (Drd1), which are expressed in motile cilia. Haploinsufficiency of the microRNA-processing gene Dgcr8 results in Drd1 elevation, which is brought about by a reduction in Drd1-targeting microRNAs miR-382-3p and miR-674-3p. Replenishing either microRNA in 22q11DS mice normalizes ciliary beating and ventricular size. Knocking down the microRNAs or deleting their seed sites on Drd1 mimicked the cilia-beating and ventricular deficits. These results suggest that the Dgcr8-miR-382-3p/miR-674-3p-Drd1 mechanism contributes to deceleration of ciliary motility and age-dependent ventricular enlargement in 22q11DS.
Collapse
Affiliation(s)
- Tae-Yeon Eom
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Seung Baek Han
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jieun Kim
- Center for In Vivo Imaging and Therapeutics, Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jay A Blundon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jing Yu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kara Anderson
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Damian B Kaminski
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Sadie Miki Sakurada
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Linda Horner
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ben Wagner
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Camenzind G Robinson
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Matthew Eicholtz
- Electrical and Electronics Systems Research Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
- Department of Computer Science, Florida Southern College, Lakeland, FL, 33801, USA
| | - Derek C Rose
- Electrical and Electronics Systems Research Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
49
|
Sandini C, Schneider M, Eliez S, Armando M. Association Between Parental Anxiety and Depression Level and Psychopathological Symptoms in Offspring With 22q11.2 Deletion Syndrome. Front Psychiatry 2020; 11:646. [PMID: 32792992 PMCID: PMC7390875 DOI: 10.3389/fpsyt.2020.00646] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
22q11.2 deletion syndrome (22q11DS) is recognized as one of the strongest genetic risk factors for the development of psychopathology, including dramatically increased prevalence of schizophrenia anxiety disorders, mood disorders, and Attention Deficit Hyperactivity Disorder (ADHD). Despite sharing a homogenous genetic deletion, the psychiatric phenotype in 22q11DS still present significant variability across subjects. The origins of such variability remain largely unclear. Levels of parental psychopathology could significantly contribute to phenotypic variability of offspring psychopathology, through mechanisms of gene x gene (GxG) and gene x environment (GxE) interactions. However, this hypothesis has not been explicitly tested to date in 22q11DS. In the present manuscript, we employed a longitudinal design to investigate bi-directional interactions of parental anxiety and depressive symptoms, estimated with Beck Depression Inventory and Beck Anxiety Inventory, and offspring level of psychopathology assessed with a combination of parentally reported Child Behavioral Checklist, Youth Self Report Questionnaire, and Structured Clinical Interviews for Prodromal Syndromes (SIPS). We tested associations in both typically developing healthy controls (HCs) (N = 88 participants; N = 131 time points) and in individuals with 22q11DS (N = 103 participants; N = 198 time points). We observed that 22q11DS individuals with higher levels of parental anxiety and depression presented significant increases in multiple forms of psychopathology, including higher internalizing and externalizing symptoms, as estimated both by parental and self-report questionnaires, along with higher negative and generalized symptoms as measured with the SIPS. Associations for positive and disorganized dimensions of the SIPS were not statistically significant. Purely longitudinal analysis pointed to bi-directional interactions of parental and child psychopathology, with marginally stronger longitudinal associations between early parental anxiety-depression and subsequent child psychopathology. Interestingly, associations between psychopathology across generations were significantly stronger in 22q11DS individuals compared to HCs. Our results show that parental levels of anxiety and depression are associated with levels of offspring psychopathology, particularly in individuals with 22q11DS. These findings point to the existence of GxG or GxE mechanisms, that should be investigated in future work. From a clinical perspective, they highlight a strong rational for the management of parental psychological well-being in 22q11DS.
Collapse
Affiliation(s)
- Corrado Sandini
- Developmental Imaging and Psychopathology lab, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Maude Schneider
- Developmental Imaging and Psychopathology lab, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva, Switzerland.,Center for Contextual Psychiatry, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Stephan Eliez
- Developmental Imaging and Psychopathology lab, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marco Armando
- Developmental Imaging and Psychopathology lab, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
50
|
Li J, Ryan SK, Deboer E, Cook K, Fitzgerald S, Lachman HM, Wallace DC, Goldberg EM, Anderson SA. Mitochondrial deficits in human iPSC-derived neurons from patients with 22q11.2 deletion syndrome and schizophrenia. Transl Psychiatry 2019; 9:302. [PMID: 31740674 PMCID: PMC6861238 DOI: 10.1038/s41398-019-0643-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/11/2019] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia (SZ) is a highly heterogeneous disorder in both its symptoms and risk factors. One of the most prevalent genetic risk factors for SZ is the hemizygous microdeletion at chromosome 22q11.2 (22q11DS) that confers a 25-fold increased risk. Six of the genes directly disrupted in 22qDS encode for mitochondrial-localizing proteins. Here, we test the hypothesis that stem cell-derived neurons from subjects with the 22q11DS and SZ have mitochondrial deficits relative to typically developing controls. Human iPSCs from four lines of affected subjects and five lines of controls were differentiated into forebrain-like excitatory neurons. In the patient group, we find significant reductions of ATP levels that appear to be secondary to reduced activity in oxidative phosphorylation complexes I and IV. Protein products of mitochondrial-encoded genes are also reduced. As one of the genes deleted in the 22q11.2 region is MRPL40, a component of the mitochondrial ribosome, we generated a heterozygous mutation of MRPL40 in a healthy control iPSC line. Relative to its isogenic control, this line shows similar deficits in mitochondrial DNA-encoded proteins, ATP level, and complex I and IV activity. These results suggest that in the 22q11DS MRPL40 heterozygosity leads to reduced mitochondria ATP production secondary to altered mitochondrial protein levels. Such defects could have profound effects on neuronal function in vivo.
Collapse
Affiliation(s)
- Jianping Li
- Department of Psychiatry, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sean K Ryan
- Department of Psychiatry, The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erik Deboer
- Mallinckrodt Pharmaceuticals, Bedminster, NJ, USA
| | - Kieona Cook
- University of Pennsylvania, Philadelphia, PA, USA
| | - Shane Fitzgerald
- Department of Psychiatry, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Herbert M Lachman
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia and Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ethan M Goldberg
- Department of Pediatrics, The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stewart A Anderson
- Department of Psychiatry, Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|