151
|
Martinez LA, Tejada-Simon MV. Pharmacological Rescue of Hippocampal Fear Learning Deficits in Fragile X Syndrome. Mol Neurobiol 2017; 55:5951-5961. [DOI: 10.1007/s12035-017-0819-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/02/2017] [Indexed: 11/28/2022]
|
152
|
Gene expression profiling in colon of mice exposed to food additive titanium dioxide (E171). Food Chem Toxicol 2017; 111:153-165. [PMID: 29128614 DOI: 10.1016/j.fct.2017.11.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/20/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
Dietary factors that may influence the risks of colorectal cancer, including specific supplements, are under investigation. Previous studies showed the capacity of food additive titanium dioxide (E171) to induce DNA damage in vitro and facilitate growth of colorectal tumours in vivo. This study aimed to investigate the molecular mechanisms behind these effects after E171 exposure. BALB/c mice were exposed by gavage to 5 mg/kgbw/day of E171 for 2, 7, 14, and 21 days. Transcriptome changes were studied by whole genome mRNA microarray analysis on the mice's distal colons. In addition, histopathological changes as well as a proliferation marker were analysed. The results showed significant gene expression changes in the olfactory/GPCR receptor family, oxidative stress, the immune system and of cancer related genes. Transcriptome analysis also identified genes that thus far have not been included in known biological pathways and can induce functional changes by interacting with other genes involved in different biological pathways. Histopathological analysis showed alteration and disruption in the normal structure of crypts inducing a hyperplastic epithelium. At cell proliferation level, no consistent increase over time was observed. These results may offer a mechanistic framework for the enhanced tumour growth after ingestion of E171 in BALB/c mice.
Collapse
|
153
|
Santini E, Huynh TN, Longo F, Koo SY, Mojica E, D'Andrea L, Bagni C, Klann E. Reducing eIF4E-eIF4G interactions restores the balance between protein synthesis and actin dynamics in fragile X syndrome model mice. Sci Signal 2017; 10:10/504/eaan0665. [PMID: 29114037 DOI: 10.1126/scisignal.aan0665] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and autism spectrum disorder. FXS is caused by silencing of the FMR1 gene, which encodes fragile X mental retardation protein (FMRP), an mRNA-binding protein that represses the translation of its target mRNAs. One mechanism by which FMRP represses translation is through its association with cytoplasmic FMRP-interacting protein 1 (CYFIP1), which subsequently sequesters and inhibits eukaryotic initiation factor 4E (eIF4E). CYFIP1 shuttles between the FMRP-eIF4E complex and the Rac1-Wave regulatory complex, thereby connecting translational regulation to actin dynamics and dendritic spine morphology, which are dysregulated in FXS model mice that lack FMRP. Treating FXS mice with 4EGI-1, which blocks interactions between eIF4E and eIF4G, a critical interaction partner for translational initiation, reversed defects in hippocampus-dependent memory and spine morphology. We also found that 4EGI-1 normalized the phenotypes of enhanced metabotropic glutamate receptor (mGluR)-mediated long-term depression (LTD), enhanced Rac1-p21-activated kinase (PAK)-cofilin signaling, altered actin dynamics, and dysregulated CYFIP1/eIF4E and CYFIP1/Rac1 interactions in FXS mice. Our findings are consistent with the idea that an imbalance in protein synthesis and actin dynamics contributes to pathophysiology in FXS mice, and suggest that targeting eIF4E may be a strategy for treating FXS.
Collapse
Affiliation(s)
- Emanuela Santini
- Center for Neural Science, New York University, New York, NY 10003, USA.,Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Thu N Huynh
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Francesco Longo
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - So Yeon Koo
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Edward Mojica
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Laura D'Andrea
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy.,Center for Human Genetics and Leuven Research Institute for Neuroscience and Disease, KU Leuven, 3000 Leuven, Belgium.,VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
154
|
Hippocampal Regulation of Postsynaptic Density Homer1 by Associative Learning. Neural Plast 2017; 2017:5959182. [PMID: 29238619 PMCID: PMC5697134 DOI: 10.1155/2017/5959182] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/18/2017] [Accepted: 10/10/2017] [Indexed: 11/18/2022] Open
Abstract
Genes involved in synaptic plasticity, particularly genes encoding postsynaptic density proteins, have been recurrently linked to psychiatric disorders including schizophrenia and autism. Postsynaptic density Homer1 proteins contribute to synaptic plasticity through the competing actions of short and long isoforms. The activity-induced expression of short Homer1 isoforms, Homer1a and Ania-3, is thought to be related to processes of learning and memory. However, the precise regulation of Homer1a and Ania-3 with different components of learning has not been investigated. Here, we used in situ hybridization to quantify short and long Homer1 expression in the hippocampus following consolidation, retrieval, and extinction of associative fear memory, using contextual fear conditioning in rats. Homer1a and Ania-3, but not long Homer1, were regulated by contextual fear learning or novelty detection, although their precise patterns of expression in hippocampal subregions were dependent on the isoform. We also show for the first time that the two short Homer1 isoforms are regulated after the retrieval and extinction of contextual fear memory, albeit with distinct temporal and spatial profiles. These findings support a role of activity-induced Homer1 isoforms in learning and memory processes in discrete hippocampal subregions and suggest that Homer1a and Ania-3 may play separable roles in synaptic plasticity.
Collapse
|
155
|
Marti AR, Patil S, Mrdalj J, Meerlo P, Skrede S, Pallesen S, Pedersen TT, Bramham CR, Grønli J. No Escaping the Rat Race: Simulated Night Shift Work Alters the Time-of-Day Variation in BMAL1 Translational Activity in the Prefrontal Cortex. Front Neural Circuits 2017; 11:70. [PMID: 29085284 PMCID: PMC5649179 DOI: 10.3389/fncir.2017.00070] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/12/2017] [Indexed: 01/26/2023] Open
Abstract
Millions of people worldwide work during the night, resulting in disturbed circadian rhythms and sleep loss. This may cause deficits in cognitive functions, impaired alertness and increased risk of errors and accidents. Disturbed circadian rhythmicity resulting from night shift work could impair brain function and cognition through disrupted synthesis of proteins involved in synaptic plasticity and neuronal function. Recently, the circadian transcription factor brain-and-muscle arnt-like protein 1 (BMAL1) has been identified as a promoter of mRNA translation initiation, the most highly regulated step in protein synthesis, through binding to the mRNA “cap”. In this study we investigated the effects of simulated shift work on protein synthesis markers. Male rats (n = 40) were exposed to forced activity, either in their rest phase (simulated night shift work) or in their active phase (simulated day shift work) for 3 days. Following the third work shift, experimental animals and time-matched undisturbed controls were euthanized (rest work at ZT12; active work at ZT0). Tissue lysates from two brain regions (prefrontal cortex, PFC and hippocampus) implicated in cognition and sleep loss, were analyzed with m7GTP (cap) pull-down to examine time-of-day variation and effects of simulated shift work on cap-bound protein translation. The results show time-of-day variation of protein synthesis markers in PFC, with increased protein synthesis at ZT12. In the hippocampus there was little difference between ZT0 and ZT12. Active phase work did not induce statistically significant changes in protein synthesis markers at ZT0 compared to time-matched undisturbed controls. Rest work, however, resulted in distinct brain-region specific changes of protein synthesis markers compared to time-matched controls at ZT12. While no changes were observed in the hippocampus, phosphorylation of cap-bound BMAL1 and its regulator S6 kinase beta-1 (S6K1) was significantly reduced in the PFC, together with significant reduction in the synaptic plasticity associated protein activity-regulatedcytoskeleton-associated protein (Arc). Our results indicate considerable time-of-day and brain-region specific variation in cap-dependent translation initiation. We concludethat simulated night shift work in rats disrupts the pathways regulating the circadian component of the translation of mRNA in the PFC, and that this may partly explain impaired waking function during night shift work.
Collapse
Affiliation(s)
- Andrea R Marti
- Bergen Stress and Sleep Group, Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Sudarshan Patil
- Department of Biomedicine, University of Bergen, Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Jelena Mrdalj
- Bergen Stress and Sleep Group, Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Peter Meerlo
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Silje Skrede
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway.,Section of Clinical Pharmacology, Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
| | - Ståle Pallesen
- Department of Psychosocial Science, University of Bergen, Bergen, Norway
| | - Torhild T Pedersen
- Bergen Stress and Sleep Group, Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Janne Grønli
- Bergen Stress and Sleep Group, Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| |
Collapse
|
156
|
Borrie SC, Brems H, Legius E, Bagni C. Cognitive Dysfunctions in Intellectual Disabilities: The Contributions of the Ras-MAPK and PI3K-AKT-mTOR Pathways. Annu Rev Genomics Hum Genet 2017; 18:115-142. [DOI: 10.1146/annurev-genom-091416-035332] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sarah C. Borrie
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Claudia Bagni
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00173 Rome, Italy
| |
Collapse
|
157
|
The non-coding RNA BC1 regulates experience-dependent structural plasticity and learning. Nat Commun 2017; 8:293. [PMID: 28819097 PMCID: PMC5561022 DOI: 10.1038/s41467-017-00311-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/19/2017] [Indexed: 11/09/2022] Open
Abstract
The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders. Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.
Collapse
|
158
|
Vidaki M, Drees F, Saxena T, Lanslots E, Taliaferro MJ, Tatarakis A, Burge CB, Wang ET, Gertler FB. A Requirement for Mena, an Actin Regulator, in Local mRNA Translation in Developing Neurons. Neuron 2017; 95:608-622.e5. [PMID: 28735747 DOI: 10.1016/j.neuron.2017.06.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/17/2017] [Accepted: 06/29/2017] [Indexed: 12/16/2022]
Abstract
During neuronal development, local mRNA translation is required for axon guidance and synaptogenesis, and dysregulation of this process contributes to multiple neurodevelopmental and cognitive disorders. However, regulation of local protein synthesis in developing axons remains poorly understood. Here, we uncover a novel role for the actin-regulatory protein Mena in the formation of a ribonucleoprotein complex that involves the RNA-binding proteins HnrnpK and PCBP1 and regulates local translation of specific mRNAs in developing axons. We find that translation of dyrk1a, a Down syndrome- and autism spectrum disorders-related gene, is dependent on Mena, both in steady-state conditions and upon BDNF stimulation. We identify hundreds of additional mRNAs that associate with the Mena complex, suggesting that it plays broader role(s) in post-transcriptional gene regulation. Our work establishes a dual role for Mena in neurons, providing a potential link between regulation of actin dynamics and local translation.
Collapse
Affiliation(s)
- Marina Vidaki
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Frauke Drees
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tanvi Saxena
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Erwin Lanslots
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew J Taliaferro
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Antonios Tatarakis
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher B Burge
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric T Wang
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Frank B Gertler
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
159
|
Synaptic Actin Dysregulation, a Convergent Mechanism of Mental Disorders? J Neurosci 2017; 36:11411-11417. [PMID: 27911743 DOI: 10.1523/jneurosci.2360-16.2016] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/25/2022] Open
Abstract
Actin polymerization governs activity-dependent modulation of excitatory synapses, including their morphology and functionality. It is clear from human genetics that neuropsychiatric and neurodevelopmental disturbances are multigenetic in nature, highlighting the need to better understand the critical neural pathways associated with these disorders and how they are altered by genetic risk alleles. One such signaling pathway that is heavily implicated by candidate genes for psychiatric and neurodevelopmental disorders are regulators of signaling to the actin cytoskeleton, suggesting that its disruption and the ensuring abnormalities of spine structures and postsynaptic complexes is a commonly affected pathway in brain disorders. This review will discuss recent experimental findings that strongly support genetic evidence linking the synaptic cytoskeleton to mental disorders, such as schizophrenia and autism spectrum disorders.
Collapse
|
160
|
Regulation of Rho GTPase proteins during spine structural plasticity for the control of local dendritic plasticity. Curr Opin Neurobiol 2017; 45:193-201. [PMID: 28709063 DOI: 10.1016/j.conb.2017.06.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/09/2017] [Indexed: 02/06/2023]
Abstract
While it is generally appreciated that learning involves the structural rearrangement of neuronal circuits, the underlying orchestration of molecular events that drives these changes is not as well understood. Recent studies on the spatiotemporal organization of synaptic signaling events have provided new insights into the biochemical underpinnings of various expressions of structural neuronal plasticity, as well as the functional consequences that emerge because of the particular behavior of the molecules involved. In particular, activity patterns of and interplay among a class of morphogenic signaling proteins, the Rho GTPases, and their downstream signals, are found to be critical for linking neuronal activity with various forms of neuronal plasticity. We review recent findings on this topic and discuss their physiological implications.
Collapse
|
161
|
Increased Training Intensity Induces Proper Membrane Localization of Actin Remodeling Proteins in the Hippocampus Preventing Cognitive Deficits: Implications for Fragile X Syndrome. Mol Neurobiol 2017; 55:4529-4542. [DOI: 10.1007/s12035-017-0666-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/19/2017] [Indexed: 10/19/2022]
|
162
|
Spatiotemporal profile of postsynaptic interactomes integrates components of complex brain disorders. Nat Neurosci 2017; 20:1150-1161. [PMID: 28671696 DOI: 10.1038/nn.4594] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/17/2017] [Indexed: 12/30/2022]
Abstract
The postsynaptic density (PSD) contains a collection of scaffold proteins used for assembling synaptic signaling complexes. However, it is not known how the core-scaffold machinery associates in protein-interaction networks or how proteins encoded by genes involved in complex brain disorders are distributed through spatiotemporal protein complexes. Here using immunopurification, proteomics and bioinformatics, we isolated 2,876 proteins across 41 in vivo interactomes and determined their protein domain composition, correlation to gene expression levels and developmental integration to the PSD. We defined clusters for enrichment of schizophrenia, autism spectrum disorders, developmental delay and intellectual disability risk factors at embryonic day 14 and adult PSD in mice. Mutations in highly connected nodes alter protein-protein interactions modulating macromolecular complexes enriched in disease risk candidates. These results were integrated into a software platform, Synaptic Protein/Pathways Resource (SyPPRes), enabling the prioritization of disease risk factors and their placement within synaptic protein interaction networks.
Collapse
|
163
|
Bozzi Y, Provenzano G, Casarosa S. Neurobiological bases of autism-epilepsy comorbidity: a focus on excitation/inhibition imbalance. Eur J Neurosci 2017; 47:534-548. [PMID: 28452083 DOI: 10.1111/ejn.13595] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/18/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorders (ASD) and epilepsy are common neurological diseases of childhood, with an estimated incidence of approximately 0.5-1% of the worldwide population. Several genetic, neuroimaging and neuropathological studies clearly showed that both ASD and epilepsy have developmental origins and a substantial degree of heritability. Most importantly, ASD and epilepsy frequently coexist in the same individual, suggesting a common neurodevelopmental basis for these disorders. Genome-wide association studies recently allowed for the identification of a substantial number of genes involved in ASD and epilepsy, some of which are mutated in syndromes presenting both ASD and epilepsy clinical features. At the cellular level, both preclinical and clinical studies indicate that the different genetic causes of ASD and epilepsy may converge to perturb the excitation/inhibition (E/I) balance, due to the dysfunction of excitatory and inhibitory circuits in various brain regions. Metabolic and immune dysfunctions, as well as environmental causes also contribute to ASD pathogenesis. Thus, an E/I imbalance resulting from neurodevelopmental deficits of multiple origins might represent a common pathogenic mechanism for both diseases. Here, we will review the most significant studies supporting these hypotheses. A deeper understanding of the molecular and cellular determinants of autism-epilepsy comorbidity will pave the way to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuri Bozzi
- Neurodevelopmental Disorders Research Group, Centre for Mind/Brain Sciences, University of Trento, via Sommarive 9, 38123, Povo, Trento, Italy.,CNR Neuroscience Institute, Pisa, Italy
| | - Giovanni Provenzano
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Simona Casarosa
- CNR Neuroscience Institute, Pisa, Italy.,Laboratory of Neural Development and Regeneration, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
164
|
Grau C, Starkovich M, Azamian MS, Xia F, Cheung SW, Evans P, Henderson A, Lalani SR, Scott DA. Xp11.22 deletions encompassing CENPVL1, CENPVL2, MAGED1 and GSPT2 as a cause of syndromic X-linked intellectual disability. PLoS One 2017; 12:e0175962. [PMID: 28414775 PMCID: PMC5393878 DOI: 10.1371/journal.pone.0175962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/03/2017] [Indexed: 12/27/2022] Open
Abstract
By searching a clinical database of over 60,000 individuals referred for array-based CNV analyses and online resources, we identified four males from three families with intellectual disability, developmental delay, hypotonia, joint hypermobility and relative macrocephaly who carried small, overlapping deletions of Xp11.22. The maximum region of overlap between their deletions spanned ~430 kb and included two pseudogenes, CENPVL1 and CENPVL2, whose functions are not known, and two protein coding genes-the G1 to S phase transition 2 gene (GSPT2) and the MAGE family member D1 gene (MAGED1). Deletions of this ~430 kb region have not been previously implicated in human disease. Duplications of GSPT2 have been documented in individuals with intellectual disability, but the phenotypic consequences of a loss of GSPT2 function have not been elucidated in humans or mouse models. Changes in MAGED1 have not been associated with intellectual disability in humans, but loss of MAGED1 function is associated with neurocognitive and neurobehavioral phenotypes in mice. In all cases, the Xp11.22 deletion was inherited from an unaffected mother. Studies performed on DNA from one of these mothers did not show evidence of skewed X-inactivation. These results suggest that deletions of an ~430 kb region on chromosome Xp11.22 that encompass CENPVL1, CENPVL2, GSPT2 and MAGED1 cause a distinct X-linked syndrome characterized by intellectual disability, developmental delay, hypotonia, joint hypermobility and relative macrocephaly. Loss of GSPT2 and/or MAGED1 function may contribute to the intellectual disability and developmental delay seen in males with these deletions.
Collapse
Affiliation(s)
- Christina Grau
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Molly Starkovich
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mahshid S. Azamian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fan Xia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Baylor Genetics, Houston, Texas, Unite States of America
| | - Sau Wai Cheung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Baylor Genetics, Houston, Texas, Unite States of America
| | - Patricia Evans
- Departments of Pediatrics and Neurology, University of Texas Southwestern Medical School, Dallas, Texas, United States of America
| | - Alex Henderson
- The Newcastle upon Tyne Hospitals, Newcastle upon Tyne, England
| | - Seema R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
165
|
Valenti D, de Bari L, Vigli D, Lacivita E, Leopoldo M, Laviola G, Vacca RA, De Filippis B. Stimulation of the brain serotonin receptor 7 rescues mitochondrial dysfunction in female mice from two models of Rett syndrome. Neuropharmacology 2017; 121:79-88. [PMID: 28419872 DOI: 10.1016/j.neuropharm.2017.04.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/21/2017] [Accepted: 04/14/2017] [Indexed: 02/07/2023]
Abstract
Rett syndrome (RTT) is a rare neurodevelopmental disorder, characterized by severe behavioral and physiological symptoms. Mutations in the methyl CpG binding protein 2 gene (MECP2) cause more than 95% of classic cases, and currently there is no cure for this devastating disorder. Recently we have demonstrated that neurobehavioral and brain molecular alterations can be rescued in a RTT mouse model, by pharmacological stimulation of the brain serotonin receptor 7 (5-HT7R). This member of the serotonin receptor family, crucially involved in the regulation of brain structural plasticity and cognitive processes, can be stimulated by systemic repeated treatment with LP-211, a brain-penetrant selective agonist. The present study extends previous findings by demonstrating that LP-211 treatment (0.25 mg/kg, once per day for 7 days) rescues mitochondrial respiratory chain impairment, oxidative phosphorylation deficiency and the reduced energy status in the brain of heterozygous female mice from two highly validated mouse models of RTT (MeCP2-308 and MeCP2-Bird mice). Moreover, LP-211 treatment completely restored the radical species overproduction by brain mitochondria in the MeCP2-308 model and partially recovered the oxidative imbalance in the more severely affected MeCP2-Bird model. These results provide the first evidence that RTT brain mitochondrial dysfunction can be rescued targeting the brain 5-HT7R and add compelling preclinical evidence of the potential therapeutic value of LP-211 as a pharmacological approach for this devastating neurodevelopmental disorder.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy.
| | - Lidia de Bari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Daniele Vigli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Enza Lacivita
- Dept. Pharmacy, University of Bari "A. Moro", via Orabona 4, 70125 Bari, Italy
| | - Marcello Leopoldo
- Dept. Pharmacy, University of Bari "A. Moro", via Orabona 4, 70125 Bari, Italy
| | - Giovanni Laviola
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
166
|
Martínez-Cerdeño V. Dendrite and spine modifications in autism and related neurodevelopmental disorders in patients and animal models. Dev Neurobiol 2017; 77:393-404. [PMID: 27390186 PMCID: PMC5219951 DOI: 10.1002/dneu.22417] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Dendrites and spines are the main neuronal structures receiving input from other neurons and glial cells. Dendritic and spine number, size, and morphology are some of the crucial factors determining how signals coming from individual synapses are integrated. Much remains to be understood about the characteristics of neuronal dendrites and dendritic spines in autism and related disorders. Although there have been many studies conducted using autism mouse models, few have been carried out using postmortem human tissue from patients. Available animal models of autism include those generated through genetic modifications and those non-genetic models of the disease. Here, we review how dendrite and spine morphology and number is affected in autism and related neurodevelopmental diseases, both in human, and genetic and non-genetic animal models of autism. Overall, data obtained from human and animal models point to a generalized reduction in the size and number, as well as an alteration of the morphology of dendrites; and an increase in spine densities with immature morphology, indicating a general spine immaturity state in autism. Additional human studies on dendrite and spine number and morphology in postmortem tissue are needed to understand the properties of these structures in the cerebral cortex of patients with autism. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.
Collapse
Affiliation(s)
- Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, UC Davis, Sacramento, California
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, North California, Sacramento, California
- MIND Institute, UC Davis School of Medicine, Sacramento, California
| |
Collapse
|
167
|
Magdalon J, Sánchez-Sánchez SM, Griesi-Oliveira K, Sertié AL. Dysfunctional mTORC1 Signaling: A Convergent Mechanism between Syndromic and Nonsyndromic Forms of Autism Spectrum Disorder? Int J Mol Sci 2017; 18:ijms18030659. [PMID: 28335463 PMCID: PMC5372671 DOI: 10.3390/ijms18030659] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/28/2022] Open
Abstract
Whereas autism spectrum disorder (ASD) exhibits striking heterogeneity in genetics and clinical presentation, dysfunction of mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway has been identified as a molecular feature common to several well-characterized syndromes with high prevalence of ASD. Additionally, recent findings have also implicated mTORC1 signaling abnormalities in a subset of nonsyndromic ASD, suggesting that defective mTORC1 pathway may be a potential converging mechanism in ASD pathology across different etiologies. However, the mechanistic evidence for a causal link between aberrant mTORC1 pathway activity and ASD neurobehavioral features varies depending on the ASD form involved. In this review, we first discuss six monogenic ASD-related syndromes, including both classical and potentially novel mTORopathies, highlighting their contribution to our understanding of the neurobiological mechanisms underlying ASD, and then we discuss existing evidence suggesting that aberrant mTORC1 signaling may also play a role in nonsyndromic ASD.
Collapse
Affiliation(s)
- Juliana Magdalon
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| | - Sandra M Sánchez-Sánchez
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil.
| | - Karina Griesi-Oliveira
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| | - Andréa L Sertié
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| |
Collapse
|
168
|
Genome-wide copy number variation analysis in a Chinese autism spectrum disorder cohort. Sci Rep 2017; 7:44155. [PMID: 28281572 PMCID: PMC5345089 DOI: 10.1038/srep44155] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/03/2017] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) describes a group of neurodevelopmental disorders with high heritability, although the underlying genetic determinants of ASDs remain largely unknown. Large-scale whole-genome studies of copy number variation in Han Chinese samples are still lacking. We performed a genome-wide copy number variation analysis of 343 ASD trios, 203 patients with sporadic cases and 988 controls in a Chinese population using Illumina genotyping platforms to identify CNVs and related genes that may contribute to ASD risk. We identified 32 rare CNVs larger than 1 Mb in 31 patients. ASD patients were found to carry a higher global burden of rare, large CNVs than controls. Recurrent de novo or case-private CNVs were found at 15q11-13, Xp22.3, 15q13.1–13.2, 3p26.3 and 2p12. The de novo 15q11–13 duplication was more prevalent in this Chinese population than in those with European ancestry. Several genes, including GRAMD2 and STAM, were implicated as novel ASD risk genes when integrating whole-genome CNVs and whole-exome sequencing data. We also identified several CNVs that include known ASD genes (SHANK3, CDH10, CSMD1) or genes involved in nervous system development (NYAP2, ST6GAL2, GRM6). Besides, our study also implicated Contactins-NYAPs-WAVE1 pathway in ASD pathogenesis. Our findings identify ASD-related CNVs in a Chinese population and implicate novel ASD risk genes and related pathway for further study.
Collapse
|
169
|
Hirayama-Kurogi M, Takizawa Y, Kunii Y, Matsumoto J, Wada A, Hino M, Akatsu H, Hashizume Y, Yamamoto S, Kondo T, Ito S, Tachikawa M, Niwa SI, Yabe H, Terasaki T, Setou M, Ohtsuki S. Downregulation of GNA13-ERK network in prefrontal cortex of schizophrenia brain identified by combined focused and targeted quantitative proteomics. J Proteomics 2017; 158:31-42. [DOI: 10.1016/j.jprot.2017.02.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 01/06/2023]
|
170
|
Salas IH, Callaerts-Vegh Z, Arranz AM, Guix FX, D’Hooge R, Esteban JA, De Strooper B, Dotti CG. Tetraspanin 6: A novel regulator of hippocampal synaptic transmission and long term plasticity. PLoS One 2017; 12:e0171968. [PMID: 28207852 PMCID: PMC5312877 DOI: 10.1371/journal.pone.0171968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/27/2017] [Indexed: 11/19/2022] Open
Abstract
Tetraspanins (Tspan) are transmembrane proteins with important scaffold and signalling functions. Deletions of Tetraspanin 6 (Tspan6) gene, a member of the tetraspanin family, have been reported in patients with Epilepsy Female-restricted with Mental Retardation (EFMR). Interestingly, mutations in Tspan7, highly homologous to Tspan6, are associated with X-linked intellectual disability, suggesting that these two proteins are important for cognition. Considering recent evidences showing that Tspan7 plays a key role in synapse development and AMPAR trafficking, we initiated the study of Tspan6 in synaptic function using a Tspan6 knock out mouse model. Here we report that hippocampal field recordings from Tspan6 knock out mice show an enhanced basal synaptic transmission and impaired long term potentiation (LTP). A normal paired-pulse facilitation response suggests that Tspan6 affects the properties of the postsynaptic rather than the presynaptic terminal. However, no changes in spine morphology or postsynaptic markers could be detected in Tspan6 KO mice compared with wild types. In addition, Tspan6 KO mice show normal locomotor behaviour and no defects in hippocampus-dependent memory tests.
Collapse
Affiliation(s)
- Isabel H. Salas
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | | | - Amaia M. Arranz
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | - Francesc X. Guix
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | - Rudi D’Hooge
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - José A. Esteban
- Centro de Biologıa Molecular ‘Severo Ochoa’ (CSIC/UAM), Madrid, Spain
| | - Bart De Strooper
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
- * E-mail: (CGD); (BDS)
| | - Carlos G. Dotti
- Centro de Biologıa Molecular ‘Severo Ochoa’ (CSIC/UAM), Madrid, Spain
- * E-mail: (CGD); (BDS)
| |
Collapse
|
171
|
Abekhoukh S, Sahin HB, Grossi M, Zongaro S, Maurin T, Madrigal I, Kazue-Sugioka D, Raas-Rothschild A, Doulazmi M, Carrera P, Stachon A, Scherer S, Drula Do Nascimento MR, Trembleau A, Arroyo I, Szatmari P, Smith IM, Milà M, Smith AC, Giangrande A, Caillé I, Bardoni B. New insights into the regulatory function of CYFIP1 in the context of WAVE- and FMRP-containing complexes. Dis Model Mech 2017; 10:463-474. [PMID: 28183735 PMCID: PMC5399562 DOI: 10.1242/dmm.025809] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 02/02/2017] [Indexed: 12/19/2022] Open
Abstract
Cytoplasmic FMRP interacting protein 1 (CYFIP1) is a candidate gene for intellectual disability (ID), autism, schizophrenia and epilepsy. It is a member of a family of proteins that is highly conserved during evolution, sharing high homology with its Drosophila homolog, dCYFIP. CYFIP1 interacts with the Fragile X mental retardation protein (FMRP, encoded by the FMR1 gene), whose absence causes Fragile X syndrome, and with the translation initiation factor eIF4E. It is a member of the WAVE regulatory complex (WRC), thus representing a link between translational regulation and the actin cytoskeleton. Here, we present data showing a correlation between mRNA levels of CYFIP1 and other members of the WRC. This suggests a tight regulation of the levels of the WRC members, not only by post-translational mechanisms, as previously hypothesized. Moreover, we studied the impact of loss of function of both CYFIP1 and FMRP on neuronal growth and differentiation in two animal models - fly and mouse. We show that these two proteins antagonize each other's function not only during neuromuscular junction growth in the fly but also during new neuronal differentiation in the olfactory bulb of adult mice. Mechanistically, FMRP and CYFIP1 modulate mTor signaling in an antagonistic manner, likely via independent pathways, supporting the results obtained in mouse as well as in fly at the morphological level. Collectively, our results illustrate a new model to explain the cellular roles of FMRP and CYFIP1 and the molecular significance of their interaction.
Collapse
Affiliation(s)
- Sabiha Abekhoukh
- Université Côte d'Azur, Nice, France.,CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France.,CNRS Associated International Laboratory (LIA) 'Neogenex', 06560 Valbonne, France
| | - H Bahar Sahin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS, UMR7104, 67400 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France
| | - Mauro Grossi
- Université Côte d'Azur, Nice, France.,CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France.,CNRS Associated International Laboratory (LIA) 'Neogenex', 06560 Valbonne, France
| | - Samantha Zongaro
- Université Côte d'Azur, Nice, France.,CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France.,CNRS Associated International Laboratory (LIA) 'Neogenex', 06560 Valbonne, France
| | - Thomas Maurin
- Université Côte d'Azur, Nice, France.,CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France.,CNRS Associated International Laboratory (LIA) 'Neogenex', 06560 Valbonne, France
| | - Irene Madrigal
- Biochemistry and Molecular Genetics Department, Hospital Clinic, 08036 Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), Barcelona, Spain.,IDIBAPS, Barcelona, Spain
| | - Daniele Kazue-Sugioka
- Université Côte d'Azur, Nice, France.,CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France.,CNRS Associated International Laboratory (LIA) 'Neogenex', 06560 Valbonne, France.,Instituto de Pesquisa Pelé Pequeno Principe, Curitiba 80250-060, Brazil
| | - Annick Raas-Rothschild
- Institute of Rare Diseases, Institute of Medical Genetics, The Chaim Sheba Medical Center, Tel Hashomer 52621, Israel
| | - Mohamed Doulazmi
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, CNRS UMR8256, IBPS, Neuroscience Paris Seine, France
| | - Pilar Carrera
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS, UMR7104, 67400 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France
| | - Andrea Stachon
- Instituto de Pesquisa Pelé Pequeno Principe, Curitiba 80250-060, Brazil
| | - Steven Scherer
- Hospital for Sick Children, Toronto, Ontario, Canada, M5G 1X8
| | | | - Alain Trembleau
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, CNRS UMR8256, IBPS, Neuroscience Paris Seine, France
| | - Ignacio Arroyo
- Center for Biomedical Research on Rare Diseases (CIBERER), Barcelona, Spain
| | - Peter Szatmari
- Centre for Addiction and Mental Health, Hospital for Sick Children, Department of Psychiatry, University of Toronto, Canada, M5G 1X8
| | - Isabel M Smith
- Departments of Pediatrics and Psychology & Neuroscience, Dalhousie University and IWK Health Centre, Halifax, Canada, B3K 6R8
| | - Montserrat Milà
- Biochemistry and Molecular Genetics Department, Hospital Clinic, 08036 Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), Barcelona, Spain.,IDIBAPS, Barcelona, Spain
| | - Adam C Smith
- Instituto de Pesquisa Pelé Pequeno Principe, Curitiba 80250-060, Brazil.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto and Program in Laboratory Medicine, University Health Network, Toronto, Canada
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS, UMR7104, 67400 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France
| | - Isabelle Caillé
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, CNRS UMR8256, IBPS, Neuroscience Paris Seine, France.,Sorbonne Paris Cité, Université Paris Diderot-Paris 7, 75013 Paris, France
| | - Barbara Bardoni
- Université Côte d'Azur, Nice, France .,CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France.,CNRS Associated International Laboratory (LIA) 'Neogenex', 06560 Valbonne, France
| |
Collapse
|
172
|
Datta D, Arion D, Roman KM, Volk DW, Lewis DA. Altered Expression of ARP2/3 Complex Signaling Pathway Genes in Prefrontal Layer 3 Pyramidal Cells in Schizophrenia. Am J Psychiatry 2017; 174:163-171. [PMID: 27523502 PMCID: PMC5288270 DOI: 10.1176/appi.ajp.2016.16020204] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Lower dendritic spine density on layer 3 pyramidal cells in the dorsolateral prefrontal cortex (DLPFC) appears to contribute to cognitive dysfunction in schizophrenia, whereas psychosis is associated with excessive dopamine release in the striatum. These findings may be related via excitatory projections from the DLPFC to the ventral mesencephalon, the location of dopamine cells projecting to the striatum. Consistent with this hypothesis, deletion of the actin-related protein-2/3 (ARP2/3) complex, which regulates the actin cytoskeleton supporting dendritic spines, produced spine loss in cortical pyramidal cells and striatal hyperdopaminergia in mice. The authors sought to determine whether the ARP2/3 complex is altered in schizophrenia. METHOD In matched pairs of schizophrenia and comparison subjects, transcript levels of ARP2/3 complex signaling pathway were assessed in laser-microdissected DLPFC layer 3 and 5 pyramidal cells and layer 3 parvalbumin interneurons, and in total DLPFC gray matter. RESULTS Transcript levels of ARP2/3 complex subunits and of nucleation promotion factors that regulate the ARP2/3 complex were significantly lower in DLPFC layer 3 and 5 pyramidal cells in schizophrenia. In contrast, these transcripts were unaltered, or only modestly changed, in parvalbumin interneurons and DLPFC gray matter. CONCLUSIONS Down-regulation of the ARP2/3 complex signaling pathway, a common final pathway for multiple signaling cascades that regulate the actin cytoskeleton, would compromise the structural stability of spines, leading to their loss. In concert with findings from deletion of the ARP2/3 complex in mice, these findings support the idea that spine deficits in the DLPFC may contribute to subcortical hyperdopaminergia in schizophrenia.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Translational Neuroscience Program, University of Pittsburgh School of Medicine,Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - Dominique Arion
- Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - Kaitlyn M. Roman
- Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - David W. Volk
- Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - David A. Lewis
- Department of Neuroscience, Translational Neuroscience Program, University of Pittsburgh School of Medicine,Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| |
Collapse
|
173
|
Schoch H, Kreibich AS, Ferri SL, White RS, Bohorquez D, Banerjee A, Port RG, Dow HC, Cordero L, Pallathra AA, Kim H, Li H, Bilker WB, Hirano S, Schultz RT, Borgmann-Winter K, Hahn CG, Feldmeyer D, Carlson GC, Abel T, Brodkin ES. Sociability Deficits and Altered Amygdala Circuits in Mice Lacking Pcdh10, an Autism Associated Gene. Biol Psychiatry 2017; 81:193-202. [PMID: 27567313 PMCID: PMC5161717 DOI: 10.1016/j.biopsych.2016.06.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/03/2016] [Accepted: 06/01/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Behavioral symptoms in individuals with autism spectrum disorder (ASD) have been attributed to abnormal neuronal connectivity, but the molecular bases of these behavioral and brain phenotypes are largely unknown. Human genetic studies have implicated PCDH10, a member of the δ2 subfamily of nonclustered protocadherin genes, in ASD. PCDH10 expression is enriched in the basolateral amygdala, a brain region implicated in the social deficits of ASD. Previous reports indicate that Pcdh10 plays a role in axon outgrowth and glutamatergic synapse elimination, but its roles in social behaviors and amygdala neuronal connectivity are unknown. We hypothesized that haploinsufficiency of Pcdh10 would reduce social approach behavior and alter the structure and function of amygdala circuits. METHODS Mice lacking one copy of Pcdh10 (Pcdh10+/-) and wild-type littermates were assessed for social approach and other behaviors. The lateral/basolateral amygdala was assessed for dendritic spine number and morphology, and amygdala circuit function was studied using voltage-sensitive dye imaging. Expression of Pcdh10 and N-methyl-D-aspartate receptor (NMDAR) subunits was assessed in postsynaptic density fractions of the amygdala. RESULTS Male Pcdh10+/- mice have reduced social approach behavior, as well as impaired gamma synchronization, abnormal spine morphology, and reduced levels of NMDAR subunits in the amygdala. Social approach deficits in Pcdh10+/- male mice were rescued with acute treatment with the NMDAR partial agonist d-cycloserine. CONCLUSIONS Our studies reveal that male Pcdh10+/- mice have synaptic and behavioral deficits, and establish Pcdh10+/- mice as a novel genetic model for investigating neural circuitry and behavioral changes relevant to ASD.
Collapse
Affiliation(s)
- Hannah Schoch
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Smilow Center for Translational Research, Room 10-170, Building 421, 3400 Civic Center Boulevard, Philadelphia, PA 19104-6168, USA
| | - Arati S. Kreibich
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Sarah L. Ferri
- Department of Biology, University of Pennsylvania, Smilow Center for Translational Research, Room 10-133, Building 421, 3400 Civic Center Boulevard, Philadelphia, PA 19104-6168, USA
| | - Rachel S. White
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Dominique Bohorquez
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Anamika Banerjee
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Russell G. Port
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Holly C. Dow
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Lucero Cordero
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Ashley A. Pallathra
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Hyong Kim
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Honghze Li
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104-6021, USA
| | - Warren B. Bilker
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104-6021, USA
| | - Shinji Hirano
- Department of Cell Biology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka 573-1010, Japan
| | - Robert T. Schultz
- Center for Autism Research, Children’s Hospital of Philadelphia, and Departments of Pediatrics and Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market Street, Philadelphia, PA 19104, USA
| | - Karin Borgmann-Winter
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA,Department of Child and Adolescent Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chang-Gyu Hahn
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Dirk Feldmeyer
- Forschungzentrum Julich, Institute of Neuroscience and Medicine, INM-2, D-52425, Julich, Germany,RWTH Aachen University, Medical School, Department of Psychiatry, Psychotherapy and Psychosomatics, D-52074 Aachen, Germany
| | - Gregory C. Carlson
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Smilow Center for Translational Research, Room 10-133, Building 421, 3400 Civic Center Boulevard, Philadelphia, PA 19104-6168, USA
| | - Edward S. Brodkin
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2220, Philadelphia, PA 19104-3403, USA
| |
Collapse
|
174
|
Anatomy and Cell Biology of Autism Spectrum Disorder: Lessons from Human Genetics. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:1-25. [PMID: 28551748 DOI: 10.1007/978-3-319-52498-6_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Until recently autism spectrum disorder (ASD) was regarded as a neurodevelopmental condition with unknown causes and pathogenesis. In the footsteps of the revolution of genome technologies and genetics, and with its high degree of heritability, ASD became the first neuropsychiatric disorder for which clues towards molecular and cellular pathogenesis were uncovered by genetic identification of susceptibility genes. Currently several hundreds of risk genes have been assigned, with a recurrence below 1% in the ASD population. The multitude and diversity of known ASD genes has extended the clinical notion that ASD comprises very heterogeneous conditions ranging from severe intellectual disabilities to mild high-functioning forms. The results of genetics have allowed to pinpoint a limited number of cellular and molecular processes likely involved in ASD including protein synthesis, signal transduction, transcription/chromatin remodelling and synaptic function all playing an essential role in the regulation of synaptic homeostasis during brain development. In this context, we highlight the role of protein synthesis as a key process in ASD pathogenesis as it might be central in synaptic deregulation and a potential target for intervention. These current insights should lead to a rational design of interventions in molecular and cellular pathways of ASD pathogenesis that may be applied to affected individuals in the future.
Collapse
|
175
|
Lin YC, Frei JA, Kilander MBC, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci 2016; 10:263. [PMID: 27909399 PMCID: PMC5112273 DOI: 10.3389/fncel.2016.00263] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/28/2016] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a range of neurological conditions that affect individuals’ ability to communicate and interact with others. People with ASD often exhibit marked qualitative difficulties in social interaction, communication, and behavior. Alterations in neurite arborization and dendritic spine morphology, including size, shape, and number, are hallmarks of almost all neurological conditions, including ASD. As experimental evidence emerges in recent years, it becomes clear that although there is broad heterogeneity of identified autism risk genes, many of them converge into similar cellular pathways, including those regulating neurite outgrowth, synapse formation and spine stability, and synaptic plasticity. These mechanisms together regulate the structural stability of neurons and are vulnerable targets in ASD. In this review, we discuss the current understanding of those autism risk genes that affect the structural connectivity of neurons. We sub-categorize them into (1) cytoskeletal regulators, e.g., motors and small RhoGTPase regulators; (2) adhesion molecules, e.g., cadherins, NCAM, and neurexin superfamily; (3) cell surface receptors, e.g., glutamatergic receptors and receptor tyrosine kinases; (4) signaling molecules, e.g., protein kinases and phosphatases; and (5) synaptic proteins, e.g., vesicle and scaffolding proteins. Although the roles of some of these genes in maintaining neuronal structural stability are well studied, how mutations contribute to the autism phenotype is still largely unknown. Investigating whether and how the neuronal structure and function are affected when these genes are mutated will provide insights toward developing effective interventions aimed at improving the lives of people with autism and their families.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Jeannine A Frei
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Michaela B C Kilander
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Wenjuan Shen
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Gene J Blatt
- Laboratory of Autism Neurocircuitry, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| |
Collapse
|
176
|
Muzar Z, Lozano R, Kolevzon A, Hagerman RJ. The neurobiology of the Prader-Willi phenotype of fragile X syndrome. Intractable Rare Dis Res 2016; 5:255-261. [PMID: 27904820 PMCID: PMC5116860 DOI: 10.5582/irdr.2016.01082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and autism, caused by a CGG expansion to greater than 200 repeats in the promoter region of FMR1 on the bottom of the X chromosome. A subgroup of individuals with FXS experience hyperphagia, lack of satiation after meals and severe obesity, this subgroup is referred to have the Prader-Willi phenotype of FXS. Prader-Willi syndrome is one of the most common genetic severe obesity disorders known and it is caused by the lack of the paternal 15q11-13 region. Affected individuals suffer from hyperphagia, lack of satiation, intellectual disability, and behavioral problems. Children with fragile X syndrome Prader-Willi phenotye and those with Prader Willi syndrome have clinical and molecular similarities reviewed here which will impact new treatment options for both disorders.
Collapse
Affiliation(s)
- Zukhrofi Muzar
- Medical Investigation of Neurodevelopmental Disorders MIND Institute, b)Department of Pediatrics, UC Davis Medical Center, Sacramento, CA, USA
- Department of Histology, Universitas Muhammadiyah Sumatera Utara (UMSU) Faculty of Medicine, Medan, North Sumatera, Indonesia
| | - Reymundo Lozano
- Seaver Autism Center for Research and Treatment, d)Departments of Genetics and Genomic Sciences, e)Psychiatry, and f)Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Address correspondence to: Dr. Reymundo Lozano, Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY 10025, USA. E-mail: Dr. Randi J. Hagerman, MIND Institute, UC Davis Health System, 2825 50th Street, Sacramento, CA 95817, USA. E-mail:
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, d)Departments of Genetics and Genomic Sciences, e)Psychiatry, and f)Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Randi J. Hagerman
- Seaver Autism Center for Research and Treatment, d)Departments of Genetics and Genomic Sciences, e)Psychiatry, and f)Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Address correspondence to: Dr. Reymundo Lozano, Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY 10025, USA. E-mail: Dr. Randi J. Hagerman, MIND Institute, UC Davis Health System, 2825 50th Street, Sacramento, CA 95817, USA. E-mail:
| |
Collapse
|
177
|
Kwan V, Meka D, White S, Hung C, Holzapfel N, Walker S, Murtaza N, Unda B, Schwanke B, Yuen R, Habing K, Milsom C, Hope K, Truant R, Scherer S, Calderon de Anda F, Singh K. DIXDC1 Phosphorylation and Control of Dendritic Morphology Are Impaired by Rare Genetic Variants. Cell Rep 2016; 17:1892-1904. [DOI: 10.1016/j.celrep.2016.10.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 09/02/2016] [Accepted: 10/14/2016] [Indexed: 10/20/2022] Open
|
178
|
Bryant CD, Yazdani N. RNA-binding proteins, neural development and the addictions. GENES BRAIN AND BEHAVIOR 2016; 15:169-86. [PMID: 26643147 DOI: 10.1111/gbb.12273] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 12/25/2022]
Abstract
Transcriptional and post-transcriptional regulation of gene expression defines the neurobiological mechanisms that bridge genetic and environmental risk factors with neurobehavioral dysfunction underlying the addictions. More than 1000 genes in the eukaryotic genome code for multifunctional RNA-binding proteins (RBPs) that can regulate all levels of RNA biogenesis. More than 50% of these RBPs are expressed in the brain where they regulate alternative splicing, transport, localization, stability and translation of RNAs during development and adulthood. Dysfunction of RBPs can exert global effects on their targetomes that underlie neurodegenerative disorders such as Alzheimer's and Parkinson's diseases as well as neurodevelopmental disorders, including autism and schizophrenia. Here, we consider the evidence that RBPs influence key molecular targets, neurodevelopment, synaptic plasticity and neurobehavioral dysfunction underlying the addictions. Increasingly well-powered genome-wide association studies in humans and mammalian model organisms combined with ever more precise transcriptomic and proteomic approaches will continue to uncover novel and possibly selective roles for RBPs in the addictions. Key challenges include identifying the biological functions of the dynamic RBP targetomes from specific cell types throughout subcellular space (e.g. the nuclear spliceome vs. the synaptic translatome) and time and manipulating RBP programs through post-transcriptional modifications to prevent or reverse aberrant neurodevelopment and plasticity underlying the addictions.
Collapse
Affiliation(s)
- C D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - N Yazdani
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
179
|
Tiwari SS, Mizuno K, Ghosh A, Aziz W, Troakes C, Daoud J, Golash V, Noble W, Hortobágyi T, Giese KP. Alzheimer-related decrease in CYFIP2 links amyloid production to tau hyperphosphorylation and memory loss. Brain 2016; 139:2751-2765. [PMID: 27524794 PMCID: PMC5035822 DOI: 10.1093/brain/aww205] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/07/2016] [Accepted: 07/01/2016] [Indexed: 11/14/2022] Open
Abstract
Characteristic features of Alzheimer's disease are memory loss, plaques resulting from abnormal processing of amyloid precursor protein (APP), and presence of neurofibrillary tangles and dystrophic neurites containing hyperphosphorylated tau. Currently, it is not known what links these abnormalities together. Cytoplasmic FMR1 interacting protein 2 (CYFIP2) has been suggested to regulate mRNA translation at synapses and this may include local synthesis of APP and alpha-calcium/calmodulin-dependent kinase II, a kinase that can phosphorylate tau. Further, CYFIP2 is part of the Wiskott-Aldrich syndrome protein-family verprolin-homologous protein complex, which has been implicated in actin polymerization at synapses, a process thought to be required for memory formation. Our previous studies on p25 dysregulation put forward the hypothesis that CYFIP2 expression is reduced in Alzheimer's disease and that this contributes to memory impairment, abnormal APP processing and tau hyperphosphorylation. Here, we tested this hypothesis. First, in post-mortem tissue CYFIP2 expression was reduced by ∼50% in severe Alzheimer's hippocampus and superior temporal gyrus when normalized to expression of a neuronal or synaptic marker protein. Interestingly, there was also a trend for decreased expression in mild Alzheimer's disease hippocampus. Second, CYFIP2 expression was reduced in old but not in young Tg2576 mice, a model of familial Alzheimer's disease. Finally, we tested the direct impact of reduced CYFIP2 expression in heterozygous null mutant mice. We found that in hippocampus this reduced expression causes an increase in APP and β-site amyloid precursor protein cleaving enzyme 1 (BACE1) protein, but not mRNA expression, and elevates production of amyloid-β42 Reduced CYFIP2 expression also increases alpha-calcium/calmodulin-dependent kinase II protein expression, and this is associated with hyperphosphorylation of tau at serine-214. The reduced expression also impairs spine maturity without affecting spine density in apical dendrites of CA1 pyramidal neurons. Furthermore, the reduced expression prevents retention of spatial memory in the water maze. Taken together, our findings indicate that reduced CYFIP2 expression triggers a cascade of change towards Alzheimer's disease, including amyloid production, tau hyperphosphorylation and memory loss. We therefore suggest that CYFIP2 could be a potential hub for targeting treatment of the disease.
Collapse
Affiliation(s)
- Sachin Suresh Tiwari
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Keiko Mizuno
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Anshua Ghosh
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Wajeeha Aziz
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Claire Troakes
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Jason Daoud
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Vidushi Golash
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Wendy Noble
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Tibor Hortobágyi
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK 2 Department of Neuropathology, Institute of Pathology, University of Debrecen, 4032 Debrecen, Hungary
| | - Karl Peter Giese
- 1 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| |
Collapse
|
180
|
Bramham CR, Jensen KB, Proud CG. Tuning Specific Translation in Cancer Metastasis and Synaptic Memory: Control at the MNK-eIF4E Axis. Trends Biochem Sci 2016; 41:847-858. [PMID: 27527252 DOI: 10.1016/j.tibs.2016.07.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 02/07/2023]
Abstract
The eukaryotic translation initiation factor (eIF) 4E, which binds to the 5'-cap of mRNA, undergoes phosphorylation on a single conserved serine, executed by the mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs). However, the functional consequences and physiological roles of MNK signalling have remained obscure. Now, new pharmacological and genetic tools have provided unprecedented insights into the function of MNKs and eIF4E phosphorylation. The studies suggest that MNKs control the translation of specific mRNAs in cancer metastasis and neuronal synaptic plasticity by a novel mechanism involving the regulation of the translational repressor, cytoplasmic fragile-X protein-interacting protein 1 (CYFIP1). These recent breakthroughs go a long way to resolving the longstanding enigma and controversy surrounding the function of the MNK-eIF4E axis in cancer cell biology and neurobiology.
Collapse
Affiliation(s)
- Clive R Bramham
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, 5009 Bergen, Norway.
| | - Kirk B Jensen
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Christopher G Proud
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
181
|
Chazeau A, Giannone G. Organization and dynamics of the actin cytoskeleton during dendritic spine morphological remodeling. Cell Mol Life Sci 2016; 73:3053-73. [PMID: 27105623 PMCID: PMC11108290 DOI: 10.1007/s00018-016-2214-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 12/18/2022]
Abstract
In the central nervous system, most excitatory post-synapses are small subcellular structures called dendritic spines. Their structure and morphological remodeling are tightly coupled to changes in synaptic transmission. The F-actin cytoskeleton is the main driving force of dendritic spine remodeling and sustains synaptic plasticity. It is therefore essential to understand how changes in synaptic transmission can regulate the organization and dynamics of actin binding proteins (ABPs). In this review, we will provide a detailed description of the organization and dynamics of F-actin and ABPs in dendritic spines and will discuss the current models explaining how the actin cytoskeleton sustains both structural and functional synaptic plasticity.
Collapse
Affiliation(s)
- Anaël Chazeau
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, UMR 5297, 33000, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, 33000, Bordeaux, France
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Grégory Giannone
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, UMR 5297, 33000, Bordeaux, France.
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, 33000, Bordeaux, France.
| |
Collapse
|
182
|
Woo YJ, Wang T, Guadalupe T, Nebel RA, Vino A, Del Bene VA, Molholm S, Ross LA, Zwiers MP, Fisher SE, Foxe JJ, Abrahams BS. A Common CYFIP1 Variant at the 15q11.2 Disease Locus Is Associated with Structural Variation at the Language-Related Left Supramarginal Gyrus. PLoS One 2016; 11:e0158036. [PMID: 27351196 PMCID: PMC4924813 DOI: 10.1371/journal.pone.0158036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 06/09/2016] [Indexed: 01/03/2023] Open
Abstract
Copy number variants (CNVs) at the Breakpoint 1 to Breakpoint 2 region at 15q11.2 (BP1-2) are associated with language-related difficulties and increased risk for developmental disorders in which language is compromised. Towards underlying mechanisms, we investigated relationships between single nucleotide polymorphisms (SNPs) across the region and quantitative measures of human brain structure obtained by magnetic resonance imaging of healthy subjects. We report an association between rs4778298, a common variant at CYFIP1, and inter-individual variation in surface area across the left supramarginal gyrus (lh.SMG), a cortical structure implicated in speech and language in independent discovery (n = 100) and validation cohorts (n = 2621). In silico analyses determined that this same variant, and others nearby, is also associated with differences in levels of CYFIP1 mRNA in human brain. One of these nearby polymorphisms is predicted to disrupt a consensus binding site for FOXP2, a transcription factor implicated in speech and language. Consistent with a model where FOXP2 regulates CYFIP1 levels and in turn influences lh.SMG surface area, analysis of publically available expression data identified a relationship between expression of FOXP2 and CYFIP1 mRNA in human brain. We propose that altered CYFIP1 dosage, through aberrant patterning of the lh.SMG, may contribute to language-related difficulties associated with BP1-2 CNVs. More generally, this approach may be useful in clarifying the contribution of individual genes at CNV risk loci.
Collapse
Affiliation(s)
- Young Jae Woo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States of America
| | - Tao Wang
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, United States of America
| | - Tulio Guadalupe
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Rebecca A. Nebel
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States of America
| | - Arianna Vino
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Victor A. Del Bene
- The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Children's Evaluation and Rehabilitation Center (CERC), Albert Einstein College of Medicine, Bronx, United States of America
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States of America
| | - Sophie Molholm
- The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Children's Evaluation and Rehabilitation Center (CERC), Albert Einstein College of Medicine, Bronx, United States of America
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States of America
| | - Lars A. Ross
- The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Children's Evaluation and Rehabilitation Center (CERC), Albert Einstein College of Medicine, Bronx, United States of America
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States of America
| | - Marcel P. Zwiers
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Simon E. Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - John J. Foxe
- The Sheryl and Daniel R. Tishman Cognitive Neurophysiology Laboratory, Children's Evaluation and Rehabilitation Center (CERC), Albert Einstein College of Medicine, Bronx, United States of America
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, United States of America
- The Cognitive Neurophysiology Laboratory, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States of America
| | - Brett S. Abrahams
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States of America
- * E-mail:
| |
Collapse
|
183
|
Abstract
UNLABELLED Copy number variations encompassing the gene encoding Cyfip1 have been associated with a variety of human diseases, including autism and schizophrenia. Here we show that juvenile mice hemizygous for Cyfip1 have altered presynaptic function, enhanced protein translation, and increased levels of F-actin. In developing hippocampus, reduced Cyfip1 levels serve to decrease paired pulse facilitation and increase miniature EPSC frequency without a change in amplitude. Higher-resolution examination shows these changes to be caused primarily by an increase in presynaptic terminal size and enhanced vesicle release probability. Short hairpin-mediated knockdown of Cyfip1 coupled with expression of mutant Cyfip1 proteins indicates that the presynaptic alterations are caused by dysregulation of the WAVE regulatory complex. Such dysregulation occurs downstream of Rac1 as acute exposure to Rac1 inhibitors rescues presynaptic responses in culture and in hippocampal slices. The data serve to highlight an early and essential role for Cyfip1 in the generation of normally functioning synapses and suggest a means by which changes in Cyfip1 levels could impact the generation of neural networks and contribute to abnormal and maladaptive behaviors. SIGNIFICANCE STATEMENT Several developmental brain disorders have been associated with gene duplications and deletions that serve to increase or decrease levels of encoded proteins. Cyfip1 is one such protein, but the role it plays in brain development is poorly understood. We asked whether decreased Cyfip1 levels altered the function of developing synapses. The data show that synapses with reduced Cyfip1 are larger and release neurotransmitter more rapidly. These effects are due to Cyfip1's role in actin polymerization and are reversed by expression of a Cyfip1 mutant protein retaining actin regulatory function or by inhibiting Rac1. Thus, Cyfip1 has a more prominent early role regulating presynaptic activity during a stage of development when activity helps to define neural pathways.
Collapse
|
184
|
Di Marino D, D'Annessa I, Tancredi H, Bagni C, Gallicchio E. A unique binding mode of the eukaryotic translation initiation factor 4E for guiding the design of novel peptide inhibitors. Protein Sci 2016; 24:1370-82. [PMID: 26013047 DOI: 10.1002/pro.2708] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/12/2015] [Accepted: 05/14/2015] [Indexed: 12/24/2022]
Abstract
The interaction between the eukaryotic translation initiation factor 4E (eIF4E) and eIF4E binding proteins (4E-BP) is a promising template for the inhibition of eIF4E and the treatment of diseases such as cancer and a spectrum of autism disorders, including the Fragile X syndrome (FXS). Here, we report an atomically detailed model of the complex between eIF4E and a peptide fragment of a 4E-BP, the cytoplasmic Fragile X interacting protein (CYFIP1). This model was generated using computer simulations with enhanced sampling from an alchemical replica exchange approach and validated using long molecular dynamics simulations. 4E-BP proteins act as post-transcriptional regulators by binding to eIF4E and preventing mRNA translation. Dysregulation of eIF4E activity has been linked to cancer, FXS, and autism spectrum disorders. Therefore, the study of the mechanism of inhibition of eIF4E by 4E-BPs is key to the development of drug therapies targeting this regulatory pathways. The results obtained in this work indicate that CYFIP1 interacts with eIF4E by an unique mode not shared by other 4E-BP proteins and elucidate the mechanism by which CYFIP1 interacts with eIF4E despite having a sequence binding motif significantly different from most 4E-BPs. Our study suggests an alternative strategy for the design of eIF4E inhibitor peptides with superior potency and specificity than currently available.
Collapse
Affiliation(s)
- Daniele Di Marino
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York, 11210
| | - Ilda D'Annessa
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Holly Tancredi
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York, 11210.,Department of Computer Science, Brooklyn College of the City University of New York, Brooklyn, New York, 11210
| | - Claudia Bagni
- VIB Center for the Biology of Disease, Leuven, Belgium.,Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), Leuven, Belgium.,Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Emilio Gallicchio
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York, 11210
| |
Collapse
|
185
|
Abstract
Although calpain was proposed to participate in synaptic plasticity and learning and memory more than 30 years ago, the mechanisms underlying its activation and the roles of different substrates have remained elusive. Recent findings have provided evidence that the two major calpain isoforms in the brain, calpain-1 and calpain-2, play opposite functions in synaptic plasticity. In particular, while calpain-1 activation is the initial trigger for certain forms of synaptic plasticity, that is, long-term potentiation, calpain-2 activation restricts the extent of plasticity. Moreover, while calpain-1 rapidly cleaves regulatory and cytoskeletal proteins, calpain-2-mediated stimulation of local protein synthesis reestablishes protein homeostasis. These findings have important implications for our understanding of learning and memory and disorders associated with impairment in these processes.
Collapse
Affiliation(s)
- Victor Briz
- 1 KU Leuven, Center for Human Genetics and Leuven Institute for Neuroscience and Disease, Leuven, Belgium
- 2 VIB Center for the Biology of Disease, Leuven, Belgium
| | - Michel Baudry
- 3 Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
186
|
Exploiting aberrant mRNA expression in autism for gene discovery and diagnosis. Hum Genet 2016; 135:797-811. [DOI: 10.1007/s00439-016-1673-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/17/2016] [Indexed: 01/09/2023]
|
187
|
Nebel RA, Zhao D, Pedrosa E, Kirschen J, Lachman HM, Zheng D, Abrahams BS. Reduced CYFIP1 in Human Neural Progenitors Results in Dysregulation of Schizophrenia and Epilepsy Gene Networks. PLoS One 2016; 11:e0148039. [PMID: 26824476 PMCID: PMC4732616 DOI: 10.1371/journal.pone.0148039] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/12/2016] [Indexed: 11/19/2022] Open
Abstract
Deletions encompassing the BP1-2 region at 15q11.2 increase schizophrenia and epilepsy risk, but only some carriers have either disorder. To investigate the role of CYFIP1, a gene within the region, we performed knockdown experiments in human neural progenitors derived from donors with 2 copies of each gene at the BP1-2 locus. RNA-seq and cellular assays determined that knockdown of CYFIP1 compromised cytoskeletal remodeling. FMRP targets and postsynaptic density genes, each implicated in schizophrenia, were significantly overrepresented among differentially expressed genes (DEGs). Schizophrenia and/or epilepsy genes, but not those associated with randomly selected disorders, were likewise significantly overrepresented. Mirroring the variable expressivity seen in deletion carriers, marked between-line differences were observed for dysregulation of disease genes. Finally, a subset of DEGs showed a striking similarity to known epilepsy genes and represents novel disease candidates. Results support a role for CYFIP1 in disease and demonstrate that disease-related biological signatures are apparent prior to neuronal differentiation.
Collapse
Affiliation(s)
- Rebecca A. Nebel
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Dejian Zhao
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jill Kirschen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Brett S. Abrahams
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
188
|
Slomnicki LP, Pietrzak M, Vashishta A, Jones J, Lynch N, Elliot S, Poulos E, Malicote D, Morris BE, Hallgren J, Hetman M. Requirement of Neuronal Ribosome Synthesis for Growth and Maintenance of the Dendritic Tree. J Biol Chem 2016; 291:5721-5739. [PMID: 26757818 DOI: 10.1074/jbc.m115.682161] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Indexed: 01/23/2023] Open
Abstract
The nucleolus serves as a principal site of ribosome biogenesis but is also implicated in various non-ribosomal functions, including negative regulation of the pro-apoptotic transcription factor p53. Although disruption of the nucleolus may trigger the p53-dependent neuronal death, neurotoxic consequences of a selective impairment of ribosome production are unclear. Here, we report that in rat forebrain neuronal maturation is associated with a remarkable expansion of ribosomes despite postnatal down-regulation of ribosomal biogenesis. In cultured rat hippocampal neurons, inhibition of the latter process by knockdowns of ribosomal proteins S6, S14, or L4 reduced ribosome content without disrupting nucleolar integrity, cell survival, and signaling responses to the neurotrophin brain-derived neurotrophic factor. Moreover, reduced general protein synthesis and/or formation of RNA stress granules suggested diminished ribosome recruitment to at least some mRNAs. Such a translational insufficiency was accompanied by impairment of brain-derived neurotrophic factor-mediated dendritic growth. Finally, RNA stress granules and smaller dendritic trees were also observed when ribosomal proteins were depleted from neurons with established dendrites. Thus, a robust ribosomal apparatus is required to carry out protein synthesis that supports dendritic growth and maintenance. Consequently, deficits of ribosomal biogenesis may disturb neurodevelopment by reducing neuronal connectivity. Finally, as stress granule formation and dendritic loss occur early in neurodegenerative diseases, disrupted homeostasis of ribosomes may initiate and/or amplify neurodegeneration-associated disconnection of neuronal circuitries.
Collapse
Affiliation(s)
- Lukasz P Slomnicki
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Maciej Pietrzak
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Aruna Vashishta
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - James Jones
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Nicholas Lynch
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Shane Elliot
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Eric Poulos
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - David Malicote
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Bridgit E Morris
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Justin Hallgren
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and
| | - Michal Hetman
- From the Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery and; Departments of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292.
| |
Collapse
|
189
|
Gabanella F, Pisani C, Borreca A, Farioli-Vecchioli S, Ciotti MT, Ingegnere T, Onori A, Ammassari-Teule M, Corbi N, Canu N, Monaco L, Passananti C, Di Certo MG. SMN affects membrane remodelling and anchoring of the protein synthesis machinery. J Cell Sci 2016; 129:804-16. [PMID: 26743087 DOI: 10.1242/jcs.176750] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/29/2015] [Indexed: 12/31/2022] Open
Abstract
Disconnection between membrane signalling and actin networks can have catastrophic effects depending on cell size and polarity. The survival motor neuron (SMN) protein is ubiquitously involved in assembly of spliceosomal small nuclear ribonucleoprotein particles. Other SMN functions could, however, affect cellular activities driving asymmetrical cell surface expansions. Genes able to mitigate SMN deficiency operate within pathways in which SMN can act, such as mRNA translation, actin network and endocytosis. Here, we found that SMN accumulates at membrane protrusions during the dynamic rearrangement of the actin filaments. In addition to localization data, we show that SMN interacts with caveolin-1, which mediates anchoring of translation machinery components. Importantly, SMN deficiency depletes the plasma membrane of ribosomes, and this correlates with the failure of fibroblasts to extend membrane protrusions. These findings strongly support a relationship between SMN and membrane dynamics. We propose that SMN could assembly translational platforms associated with and governed by the plasma membrane. This activity could be crucial in cells that have an exacerbated interdependence of membrane remodelling and local protein synthesis.
Collapse
Affiliation(s)
- Francesca Gabanella
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Cinzia Pisani
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Antonella Borreca
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Stefano Farioli-Vecchioli
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Maria Teresa Ciotti
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy European Brain Research Institute (EBRI) Rita Levi-Montalcini, Rome 00143, Italy
| | - Tiziano Ingegnere
- Department of Ecological and Biological Sciences, Tuscia University, Viterbo 01100, Italy
| | - Annalisa Onori
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Martine Ammassari-Teule
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Nicoletta Corbi
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Nadia Canu
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy Department of System Medicine, University of 'Tor Vergata', Rome 00137, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome 00185, Italy
| | - Claudio Passananti
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Maria Grazia Di Certo
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| |
Collapse
|
190
|
Huguet G, Benabou M, Bourgeron T. The Genetics of Autism Spectrum Disorders. RESEARCH AND PERSPECTIVES IN ENDOCRINE INTERACTIONS 2016. [DOI: 10.1007/978-3-319-27069-2_11] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
191
|
From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat Rev Neurosci 2015; 16:551-63. [PMID: 26289574 DOI: 10.1038/nrn3992] [Citation(s) in RCA: 649] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genetics studies of autism spectrum disorder (ASD) have identified several risk genes that are key regulators of synaptic plasticity. Indeed, many of the risk genes that have been linked to these disorders encode synaptic scaffolding proteins, receptors, cell adhesion molecules or proteins that are involved in chromatin remodelling, transcription, protein synthesis or degradation, or actin cytoskeleton dynamics. Changes in any of these proteins can increase or decrease synaptic strength or number and, ultimately, neuronal connectivity in the brain. In addition, when deleterious mutations occur, inefficient genetic buffering and impaired synaptic homeostasis may increase an individual's risk for ASD.
Collapse
|
192
|
Conte F, Oti M, Dixon J, Carels CEL, Rubini M, Zhou H. Systematic analysis of copy number variants of a large cohort of orofacial cleft patients identifies candidate genes for orofacial clefts. Hum Genet 2015; 135:41-59. [PMID: 26561393 PMCID: PMC4698300 DOI: 10.1007/s00439-015-1606-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022]
Abstract
Orofacial clefts (OFCs) represent a large fraction of human birth defects and are one of the most common phenotypes affected by large copy number variants (CNVs). Due to the limited number of CNV patients in individual centers, CNV analyses of a large number of OFC patients are challenging. The present study analyzed 249 genomic deletions and 226 duplications from a cohort of 312 OFC patients reported in two publicly accessible databases of chromosome imbalance and phenotype in humans, DECIPHER and ECARUCA. Genomic regions deleted or duplicated in multiple patients were identified, and genes in these overlapping CNVs were prioritized based on the number of genes encompassed by the region and gene expression in embryonic mouse palate. Our analyses of these overlapping CNVs identified two genes known to be causative for human OFCs, SATB2 and MEIS2, and 12 genes (DGCR6, FGF2, FRZB, LETM1, MAPK3, SPRY1, THBS1, TSHZ1, TTC28, TULP4, WHSC1, WHSC2) that are associated with OFC or orofacial development. Additionally, we report 34 deleted and 24 duplicated genes that have not previously been associated with OFCs but are associated with the BMP, MAPK and RAC1 pathways. Statistical analyses show that the high number of overlapping CNVs is not due to random occurrence. The identified genes are not located in highly variable genomic regions in healthy populations and are significantly enriched for genes that are involved in orofacial development. In summary, we report a CNV analysis pipeline of a large cohort of OFC patients and identify novel candidate OFC genes.
Collapse
Affiliation(s)
- Federica Conte
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands.,Medical Genetic Unit, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Martin Oti
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | - Jill Dixon
- Faculty of Medical and Human Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Carine E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michele Rubini
- Medical Genetic Unit, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy.
| | - Huiqing Zhou
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands. .,Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
193
|
Topol A, English JA, Flaherty E, Rajarajan P, Hartley BJ, Gupta S, Desland F, Zhu S, Goff T, Friedman L, Rapoport J, Felsenfeld D, Cagney G, Mackay-Sim A, Savas JN, Aronow B, Fang G, Zhang B, Cotter D, Brennand KJ. Increased abundance of translation machinery in stem cell-derived neural progenitor cells from four schizophrenia patients. Transl Psychiatry 2015; 5:e662. [PMID: 26485546 PMCID: PMC4930118 DOI: 10.1038/tp.2015.118] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 12/17/2022] Open
Abstract
The genetic and epigenetic factors contributing to risk for schizophrenia (SZ) remain unresolved. Here we demonstrate, for the first time, perturbed global protein translation in human-induced pluripotent stem cell (hiPSC)-derived forebrain neural progenitor cells (NPCs) from four SZ patients relative to six unaffected controls. We report increased total protein levels and protein synthesis, together with two independent sets of quantitative mass spectrometry evidence indicating markedly increased levels of ribosomal and translation initiation and elongation factor proteins, in SZ hiPSC NPCs. We posit that perturbed levels of global protein synthesis in SZ hiPSC NPCs represent a novel post-transcriptional mechanism that might contribute to disease progression.
Collapse
Affiliation(s)
- A Topol
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J A English
- Royal College of Surgeons in Ireland, Beaumont Hospital, Beaumont, Dublin, Ireland
| | - E Flaherty
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - P Rajarajan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - B J Hartley
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S Gupta
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - F Desland
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S Zhu
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - T Goff
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Friedman
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - J Rapoport
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - D Felsenfeld
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - G Cagney
- UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - A Mackay-Sim
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | - J N Savas
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - B Aronow
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - G Fang
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - B Zhang
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D Cotter
- Royal College of Surgeons in Ireland, Beaumont Hospital, Beaumont, Dublin, Ireland
| | - K J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
194
|
Parental origin impairment of synaptic functions and behaviors in cytoplasmic FMRP interacting protein 1 (Cyfip1) deficient mice. Brain Res 2015; 1629:340-50. [PMID: 26474913 DOI: 10.1016/j.brainres.2015.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 09/01/2015] [Accepted: 10/06/2015] [Indexed: 11/30/2022]
Abstract
CYFIP1 maps to the interval between proximal breakpoint 1 (BP1) and breakpoint 2 (BP2) of chromosomal 15q11-q13 deletions that are implicated in the Angelman (AS) and Prader-Willi syndrome (PWS). There is only one breakpoint (BP3) at the distal end of deletion. CYFIP1 is deleted in AS patients with the larger class I deletion (BP1 to BP3) and the neurological presentations in these patients are more severe than that of patients with class II (BP2 to BP3) deletion. The haploinsufficiency of CYFIP1 is hypothesized to contribute to more severe clinical presentations in class I AS patients. The expression of CYFIP1 is suggested to be bi-allelic in literature but the possibility of parental origin of expression is not completely excluded. We generated and characterized Cyfip1 mutant mice. Homozygous Cyfip1 mice were early embryonic lethal. However, there was a parental origin specific effect between paternal Cyfip1 deficiency (m+/p-) and maternal deficiency (m-/p+) on both synaptic transmissions and behaviors in hippocampal CA1 synapses despite no evidence supporting the parental origin difference for the expression. Both m-/p+ and m+/p- showed the impaired input-output response and paired-pulse facilitation. While the long term-potentiation and group I mGluR mediated long term depression induced by DHPG was not different between Cyfip1 m-/p+ and m+/p- mice, the initial DHPG induced response was significantly enhanced in m-/p+ but not in m+/p- mice. m+/p- but not m-/p+ mice displayed increased freezing in cued fear conditioning and abnormal transitions in zero-maze test. The impaired synaptic transmission and behaviors in haploinsufficiency of Cyfip1 mice provide the evidence supporting the role of CYFIP1 modifying the clinical presentation of class I AS patients and in human neuropsychiatric disorders.
Collapse
|
195
|
Induction of Interleukin-1β by Human Immunodeficiency Virus-1 Viral Proteins Leads to Increased Levels of Neuronal Ferritin Heavy Chain, Synaptic Injury, and Deficits in Flexible Attention. J Neurosci 2015. [PMID: 26203149 DOI: 10.1523/jneurosci.4403-14.2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Synaptodendritic pruning and alterations in neurotransmission are the main underlying causes of HIV-associated neurocognitive disorders (HAND). Our studies in humans and nonhuman primates indicated that the protein ferritin heavy chain (FHC) is a critical player in neuronal changes and ensuing cognitive deficit observed in these patients. Here we focus on the effect of HIV proteins and inflammatory cytokines implicated in HAND on neuronal FHC levels, dendritic changes, and neurocognitive behavior. In two well characterized models of HAND (HIV transgenic and gp120-treated rats), we report reductions in spine density and dendritic branches in prefrontal cortex pyramidal neurons compared with age-matched controls. FHC brain levels are elevated in these animals, which also show deficits in reversal learning. Moreover, IL-1β, TNF-α, and HIV gp120 upregulate FHC in rat cortical neurons. However, although the inflammatory cytokines directly altered neuronal FHC, gp120 only caused significant FHC upregulation in neuronal/glial cocultures, suggesting that glia are necessary for sustained elevation of neuronal FHC by the viral protein. Although the envelope protein induced secretion of IL-1β and TNF-α in cocultures, TNF-α blockade did not affect gp120-mediated induction of FHC. Conversely, studies with an IL-1β neutralizing antibody or specific IL-1 receptor antagonist revealed the primary involvement of IL-1β in gp120-induced FHC changes. Furthermore, silencing of neuronal FHC abrogates the effect of gp120 on spines, and spine density correlates negatively with FHC levels or cognitive deficit. These results demonstrate that viral and host components of HIV infection increase brain expression of FHC, leading to cellular and functional changes, and point to IL-1β-targeted strategies for prevention of these alterations. Significance statement: This work demonstrates the key role of the cytokine IL-1β in the regulation of a novel intracellular mediator [i.e., the protein ferritin heavy chain (FHC)] of HIV-induced dendritic damage and the resulting neurocognitive impairment. This is also the first study that systematically investigates dendritic damage in layer II/III prefrontal cortex neurons of two different non-infectious models of HIV-associated neurocognitive disorders (HAND) and reveals a precise correlation of these structural changes with specific biochemical and functional alterations also reported in HIV patients. Overall, these data suggest that targeting the IL-1β-dependent FHC increase may represent a valid strategy for neuroprotective adjuvant therapies in HAND.
Collapse
|
196
|
Reduced protein synthesis in schizophrenia patient-derived olfactory cells. Transl Psychiatry 2015; 5:e663. [PMID: 26485547 PMCID: PMC4930119 DOI: 10.1038/tp.2015.119] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/16/2015] [Accepted: 07/08/2015] [Indexed: 11/16/2022] Open
Abstract
Human olfactory neurosphere-derived (ONS) cells have the potential to provide novel insights into the cellular pathology of schizophrenia. We used discovery-based proteomics and targeted functional analyses to reveal reductions in 17 ribosomal proteins, with an 18% decrease in the total ribosomal signal intensity in schizophrenia-patient-derived ONS cells. We quantified the rates of global protein synthesis in vitro and found a significant reduction in the rate of protein synthesis in schizophrenia patient-derived ONS cells compared with control-derived cells. Protein synthesis rates in fibroblast cell lines from the same patients did not differ, suggesting cell type-specific effects. Pathway analysis of dysregulated proteomic and transcriptomic data sets from these ONS cells converged to highlight perturbation of the eIF2α, eIF4 and mammalian target of rapamycin (mTOR) translational control pathways, and these pathways were also implicated in an independent induced pluripotent stem cell-derived neural stem model, and cohort, of schizophrenia patients. Analysis in schizophrenia genome-wide association data from the Psychiatric Genetics Consortium specifically implicated eIF2α regulatory kinase EIF2AK2, and confirmed the importance of the eIF2α, eIF4 and mTOR translational control pathways at the level of the genome. Thus, we integrated data from proteomic, transcriptomic, and functional assays from schizophrenia patient-derived ONS cells with genomics data to implicate dysregulated protein synthesis for the first time in schizophrenia.
Collapse
|
197
|
Richter JD, Bassell GJ, Klann E. Dysregulation and restoration of translational homeostasis in fragile X syndrome. Nat Rev Neurosci 2015; 16:595-605. [PMID: 26350240 DOI: 10.1038/nrn4001] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fragile X syndrome (FXS), the most-frequently inherited form of intellectual disability and the most-prevalent single-gene cause of autism, results from a lack of fragile X mental retardation protein (FMRP), an RNA-binding protein that acts, in most cases, to repress translation. Multiple pharmacological and genetic manipulations that target receptors, scaffolding proteins, kinases and translational control proteins can rescue neuronal morphology, synaptic function and behavioural phenotypes in FXS model mice, presumably by reducing excessive neuronal translation to normal levels. Such rescue strategies might also be explored in the future to identify the mRNAs that are critical for FXS pathophysiology.
Collapse
Affiliation(s)
- Joel D Richter
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01545, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York City, New York 10003, USA
| |
Collapse
|
198
|
Maruani A, Huguet G, Beggiato A, ElMaleh M, Toro R, Leblond CS, Mathieu A, Amsellem F, Lemière N, Verloes A, Leboyer M, Gillberg C, Bourgeron T, Delorme R. 11q24.2-25 micro-rearrangements in autism spectrum disorders: Relation to brain structures. Am J Med Genet A 2015; 167A:3019-30. [DOI: 10.1002/ajmg.a.37345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 08/12/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Anna Maruani
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
| | - Guillaume Huguet
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Anita Beggiato
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
| | - Monique ElMaleh
- Department of Radiology; Robert Debré Hospital, APHP; Paris France
| | - Roberto Toro
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Claire S. Leblond
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Alexandre Mathieu
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | | | - Nathalie Lemière
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
| | - Alain Verloes
- Department of Human Genetics; Robert Debré Hospital, APHP; Paris France
| | - Marion Leboyer
- INSERM U955, Team 15; Faculty of Medicine; Creteil France
- Department of Adult Psychiatry; Henri Mondor-Albert Chenevier Hospitals AP-HP; Creteil France
- Fondation FondaMental; French National Science Foundation; Creteil France
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre; Gothenburg University; Göteborg Sweden
- Saint George's Hospital Medical School; London United Kingdom
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Diderot Paris 7; University Paris; Paris France
| | - Richard Delorme
- Human Genetics and Cognitive Functions; Institut Pasteur; Paris France
- CNRS URA 2182; Institut Pasteur; Paris France
- Department of Child Psychiatry; Robert Debré Hospital, APHP; Paris France
- Fondation FondaMental; French National Science Foundation; Creteil France
| |
Collapse
|
199
|
Oguro-Ando A, Rosensweig C, Herman E, Nishimura Y, Werling D, Bill BR, Berg JM, Gao F, Coppola G, Abrahams BS, Geschwind DH. Increased CYFIP1 dosage alters cellular and dendritic morphology and dysregulates mTOR. Mol Psychiatry 2015; 20:1069-78. [PMID: 25311365 PMCID: PMC4409498 DOI: 10.1038/mp.2014.124] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 07/18/2014] [Accepted: 08/21/2014] [Indexed: 12/22/2022]
Abstract
Rare maternally inherited duplications at 15q11-13 are observed in ~1% of individuals with an autism spectrum disorder (ASD), making it among the most common causes of ASD. 15q11-13 comprises a complex region, and as this copy number variation encompasses many genes, it is important to explore individual genotype-phenotype relationships. Cytoplasmic FMR1-interacting protein 1 (CYFIP1) is of particular interest because of its interaction with Fragile X mental retardation protein (FMRP), its upregulation in transformed lymphoblastoid cell lines from patients with duplications at 15q11-13 and ASD and the presence of smaller overlapping deletions of CYFIP1 in patients with schizophrenia and intellectual disability. Here, we confirm that CYFIP1 is upregulated in transformed lymphoblastoid cell lines and demonstrate its upregulation in the post-mortem brain from 15q11-13 duplication patients for the first time. To investigate how increased CYFIP1 dosage might predispose to neurodevelopmental disease, we studied the consequence of its overexpression in multiple systems. We show that overexpression of CYFIP1 results in morphological abnormalities including cellular hypertrophy in SY5Y cells and differentiated mouse neuronal progenitors. We validate these results in vivo by generating a BAC transgenic mouse, which overexpresses Cyfip1 under the endogenous promotor, observing an increase in the proportion of mature dendritic spines and dendritic spine density. Gene expression profiling on embryonic day 15 suggested the dysregulation of mammalian target of rapamycin (mTOR) signaling, which was confirmed at the protein level. Importantly, similar evidence of mTOR-related dysregulation was seen in brains from 15q11-13 duplication patients with ASD. Finally, treatment of differentiated mouse neuronal progenitors with an mTOR inhibitor (rapamycin) rescued the morphological abnormalities resulting from CYFIP1 overexpression. Together, these data show that CYFIP1 overexpression results in specific cellular phenotypes and implicate modulation by mTOR signaling, further emphasizing its role as a potential convergent pathway in some forms of ASD.
Collapse
Affiliation(s)
- A Oguro-Ando
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
,Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - C Rosensweig
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - E Herman
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - Y Nishimura
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - D Werling
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - BR Bill
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - JM Berg
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - F Gao
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - G Coppola
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
,Semel Institute, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South, Los Angeles, CA 90095-1761
| | - BS Abrahams
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
| | - DH Geschwind
- Programs in Neurogenetics, Department of. Neurology and Program in Neurobehavioral Genetics and Center for Autism Research and Treatment, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South Los Angeles, CA 90095-1761
,Dept. of Human Genetics, David Geffen School of Medicine, University of California at Los Angeles, 2309 Gonda Bldg, 695 Charles E. Young Dr. South, Los Angeles, CA 90095-1761
,
| |
Collapse
|
200
|
Huang Y. Up-regulated cytoplasmic FMRP-interacting protein 1 in intractable temporal lobe epilepsy patients and a rat model. Int J Neurosci 2015; 126:542-551. [PMID: 26000921 DOI: 10.3109/00207454.2015.1038711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cytoplasmic FMRP-interacting protein 1 (CYFIP1) is a multifunctional protein which expresses highly at excitatory synapses and can locally regulate actin cytoskeletal dynamics, spine morphology and synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor lateral diffusion. Altered synaptic actin plays a role in the pathogenesis of epilepsy. The aim of this study was to investigate the expression pattern of CYFIP1 in temporal lobe epilepsy (TLE). Protein and mRNA expression levels were compared in temporal lobe tissue from patients with TLE versus trauma patients without TLE using quantitative real-time polymerase chain reaction (qRT-PCR), double-label immunofluorescence and Western blot analysis. We have further determined the expression pattern of Cyfip1 mRNA and protein in the hippocampus and adjacent cortex of a common rat model of TLE, lithium-pilocarpine treatment, compared to control rats. CYFIP1 expression was significantly up-regulated in the temporal neocortex of patients with intractable TLE and pilocarpine-treated rats compared to control groups. CYFIP1 localizes to the cytoplasm of neurons, and is not expressed in the astrocytes. Furthermore, CYFIP1 expression levels increased significantly in the two months after pilocarpine treatment, which corresponds to the period of epileptogenesis. Thus, our results indicate that CYFIP1 may be involved in the pathogenesis of TLE.
Collapse
Affiliation(s)
- Yunyi Huang
- a Department of Neurology, The Second Affiliated Hospital , Chongqing Medical University , Chongqing , China
| |
Collapse
|