1
|
Falkovich R, Aryal S, Wang J, Sheng M, Bathe M. Synaptic composition, activity, mRNA translation and dynamics in combined single-synapse profiling using multimodal imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620504. [PMID: 39554017 PMCID: PMC11565908 DOI: 10.1101/2024.10.28.620504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The function of neuronal circuits, and its perturbation by psychoactive molecules or disease-associated genetic variants, is governed by the interplay between synapse activity and synaptic protein localization and synthesis across a heterogeneous synapse population. Here, we combine in situ measurement of synaptic multiprotein compositions and activation states, synapse activity in calcium traces or glutamate spiking, and local translation of specific genes, across the same individual synapses. We demonstrate how this high-dimensional data enables identification of interdependencies in the multiprotein-activity network, and causal dissection of complex synaptic phenotypes in disease-relevant chemical and genetic NMDAR loss of function that translate in vivo . We show how this method generalizes to other subcellular systems by deriving mitochondrial protein networks, and, using support vector machines, its value in overcoming animal variability in phenotyping. Integrating multiple synapse information modalities enables deep structure-function characterization of synapse populations and their responses to genetic and chemical perturbations.
Collapse
|
2
|
Samardžija B, Petrović M, Zaharija B, Medija M, Meštrović A, Bradshaw NJ, Filošević Vujnović A, Andretić Waldowski R. Transgenic Drosophila melanogaster Carrying a Human Full-Length DISC1 Construct (UAS- hflDISC1) Showing Effects on Social Interaction Networks. Curr Issues Mol Biol 2024; 46:8526-8549. [PMID: 39194719 DOI: 10.3390/cimb46080502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Disrupted in Schizophrenia 1 (DISC1) is a scaffold protein implicated in major mental illnesses including schizophrenia, with a significant negative impact on social life. To investigate if DISC1 affects social interactions in Drosophila melanogaster, we created transgenic flies with second or third chromosome insertions of the human full-length DISC1 (hflDISC1) gene fused to a UAS promotor (UAS-hflDISC1). Initial characterization of the insertion lines showed unexpected endogenous expression of the DISC1 protein that led to various behavioral and neurochemical phenotypes. Social interaction network (SIN) analysis showed altered social dynamics and organizational structures. This was in agreement with the altered levels of the locomotor activity of individual flies monitored for 24 h. Together with a decreased ability to climb vertical surfaces, the observed phenotypes indicate altered motor functions that could be due to a change in the function of the motor neurons and/or central brain. The changes in social behavior and motor function suggest that the inserted hflDISC1 gene influences nervous system functioning that parallels symptoms of DISC1-related mental diseases in humans. Furthermore, neurochemical analyses of transgenic lines revealed increased levels of hydrogen peroxide and decreased levels of glutathione, indicating an impact of DISC1 on the dynamics of redox regulation, similar to that reported in transgenic mammals. Future studies are needed to address the localization of DISC1 expression and to address how the redox parameter changes correlate with the observed behavioral changes.
Collapse
Affiliation(s)
- Bobana Samardžija
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Milan Petrović
- Faculty of Informatics and Digital Technologies, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Beti Zaharija
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Marta Medija
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Ana Meštrović
- Faculty of Informatics and Digital Technologies, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Nicholas J Bradshaw
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Ana Filošević Vujnović
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| | - Rozi Andretić Waldowski
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51 000 Rijeka, Croatia
| |
Collapse
|
3
|
Cohen H, Matar MA, Todder D, Cohen C, Zohar J, Hawlena H, Abramsky Z. Sounds of danger and post-traumatic stress responses in wild rodents: ecological validity of a translational model of post-traumatic stress disorder. Mol Psychiatry 2023; 28:4719-4728. [PMID: 37674017 PMCID: PMC10914612 DOI: 10.1038/s41380-023-02240-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023]
Abstract
In the wild, animals face a highly variable world full of predators. Most predator attacks are unsuccessful, and the prey survives. According to the conventional perspective, the fear responses elicited by predators are acute and transient in nature. However, the long-term, non-lethal effects of predator exposure on prey behavioral stress sequelae, such as anxiety and post-traumatic symptoms, remain poorly understood. Most experiments on animal models of anxiety-related behavior or post-traumatic stress disorder have been carried out using commercial strains of rats and mice. A fundamental question is whether laboratory rodents appropriately express the behavioral responses of wild species in their natural environment; in other words, whether behavioral responses to stress observed in the laboratory can be generalized to natural behavior. To further elucidate the relative contributions of the natural selection pressures influences, this study investigated the bio-behavioral and morphological effects of auditory predator cues (owl territorial calls) in males and females of three wild rodent species in a laboratory set-up: Acomys cahirinus; Gerbillus henleyi; and Gerbillus gerbillus. Our results indicate that owl territorial calls elicited not only "fight or flight" behavioral responses but caused PTSD-like behavioral responses in wild rodents that have never encountered owls in nature and could cause, in some individuals, enduring physiological and morphological responses that parallel those seen in laboratory rodents or traumatized people. In all rodent species, the PTSD phenotype was characterized by a blunting of fecal cortisol metabolite response early after exposure and by a lower hypothalamic orexin-A level and lower total dendritic length and number in the dentate gyrus granule cells eight days after predator exposure. Phenotypically, this refers to a significant functional impairment that could affect reproduction and survival and thus fitness and population dynamics.
Collapse
Affiliation(s)
- Hagit Cohen
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel & Ministry of Health, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Beer-Sheva, Israel.
- Department of Psychology, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Michael A Matar
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel & Ministry of Health, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Beer-Sheva, Israel
| | - Doron Todder
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel & Ministry of Health, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Beer-Sheva, Israel
| | - Carmit Cohen
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel & Ministry of Health, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Beer-Sheva, Israel
| | - Joseph Zohar
- Post-Trauma Center, Sheba Medical Center, Tel Aviv University, Tel Aviv, 52621, Israel
| | - Hadas Hawlena
- Mitrani Department of Desert Ecology, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Midreshet Ben-Gurion Israel, Sde Boker, 8499000, Israel
| | - Zvika Abramsky
- Department of Life Sciences and Ramon Science Center, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| |
Collapse
|
4
|
Edwards N, Combrinck C, McCaughey-Chapman A, Connor B. Directly reprogrammed fragile X syndrome dorsal forebrain precursor cells generate cortical neurons exhibiting impaired neuronal maturation. Front Cell Neurosci 2023; 17:1254412. [PMID: 37810261 PMCID: PMC10552551 DOI: 10.3389/fncel.2023.1254412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction The neurodevelopmental disorder fragile X syndrome (FXS) is the most common monogenic cause of intellectual disability associated with autism spectrum disorder. Inaccessibility to developing human brain cells is a major barrier to studying FXS. Direct-to-neural precursor reprogramming provides a unique platform to investigate the developmental profile of FXS-associated phenotypes throughout neural precursor and neuron generation, at a temporal resolution not afforded by post-mortem tissue and in a patient-specific context not represented in rodent models. Direct reprogramming also circumvents the protracted culture times and low efficiency of current induced pluripotent stem cell strategies. Methods We have developed a chemically modified mRNA (cmRNA) -based direct reprogramming protocol to generate dorsal forebrain precursors (hiDFPs) from FXS patient-derived fibroblasts, with subsequent differentiation to glutamatergic cortical neurons and astrocytes. Results We observed differential expression of mature neuronal markers suggesting impaired neuronal development and maturation in FXS- hiDFP-derived neurons compared to controls. FXS- hiDFP-derived cortical neurons exhibited dendritic growth and arborization deficits characterized by reduced neurite length and branching consistent with impaired neuronal maturation. Furthermore, FXS- hiDFP-derived neurons exhibited a significant decrease in the density of pre- and post- synaptic proteins and reduced glutamate-induced calcium activity, suggesting impaired excitatory synapse development and functional maturation. We also observed a reduced yield of FXS- hiDFP-derived neurons with a significant increase in FXS-affected astrocytes. Discussion This study represents the first reported derivation of FXS-affected cortical neurons following direct reprogramming of patient fibroblasts to dorsal forebrain precursors and subsequently neurons that recapitulate the key molecular hallmarks of FXS as it occurs in human tissue. We propose that direct to hiDFP reprogramming provides a unique platform for further study into the pathogenesis of FXS as well as the identification and screening of new drug targets for the treatment of FXS.
Collapse
Affiliation(s)
| | | | | | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Falkovich R, Danielson EW, Perez de Arce K, Wamhoff EC, Strother J, Lapteva AP, Sheng M, Cottrell JR, Bathe M. A synaptic molecular dependency network in knockdown of autism- and schizophrenia-associated genes revealed by multiplexed imaging. Cell Rep 2023; 42:112430. [PMID: 37099425 DOI: 10.1016/j.celrep.2023.112430] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/29/2023] [Accepted: 04/08/2023] [Indexed: 04/27/2023] Open
Abstract
The complex functions of neuronal synapses depend on their tightly interconnected protein network, and their dysregulation is implicated in the pathogenesis of autism spectrum disorders and schizophrenia. However, it remains unclear how synaptic molecular networks are altered biochemically in these disorders. Here, we apply multiplexed imaging to probe the effects of RNAi knockdown of 16 autism- and schizophrenia-associated genes on the simultaneous joint distribution of 10 synaptic proteins, observing several protein composition phenotypes associated with these risk genes. We apply Bayesian network analysis to infer hierarchical dependencies among eight excitatory synaptic proteins, yielding predictive relationships that can only be accessed with single-synapse, multiprotein measurements performed simultaneously in situ. Finally, we find that central features of the network are affected similarly across several distinct gene knockdowns. These results offer insight into the convergent molecular etiology of these widespread disorders and provide a general framework to probe subcellular molecular networks.
Collapse
Affiliation(s)
- Reuven Falkovich
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric W Danielson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Karen Perez de Arce
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eike-C Wamhoff
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Juliana Strother
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna P Lapteva
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey R Cottrell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark Bathe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School Initiative for RNA Medicine, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
6
|
Cote JL, Vander PB, Ellis M, Cline JM, Svezhova N, Doche ME, Maures TJ, Choudhury TA, Kong S, Klaft OGJ, Joe RM, Argetsinger LS, Carter-Su C. The nucleolar δ isoform of adapter protein SH2B1 enhances morphological complexity and function of cultured neurons. J Cell Sci 2022; 135:jcs259179. [PMID: 35019135 PMCID: PMC8918807 DOI: 10.1242/jcs.259179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/22/2021] [Indexed: 11/20/2022] Open
Abstract
The adapter protein SH2B1 is recruited to neurotrophin receptors, including TrkB (also known as NTRK2), the receptor for brain-derived neurotrophic factor (BDNF). Herein, we demonstrate that the four alternatively spliced isoforms of SH2B1 (SH2B1α-SH2B1δ) are important determinants of neuronal architecture and neurotrophin-induced gene expression. Primary hippocampal neurons from Sh2b1-/- [knockout (KO)] mice exhibit decreased neurite complexity and length, and BDNF-induced expression of the synapse-related immediate early genes Egr1 and Arc. Reintroduction of each SH2B1 isoform into KO neurons increases neurite complexity; the brain-specific δ isoform also increases total neurite length. Human obesity-associated variants, when expressed in SH2B1δ, alter neurite complexity, suggesting that a decrease or increase in neurite branching may have deleterious effects that contribute to the severe childhood obesity and neurobehavioral abnormalities associated with these variants. Surprisingly, in contrast to SH2B1α, SH2B1β and SH2B1γ, which localize primarily in the cytoplasm and plasma membrane, SH2B1δ resides primarily in nucleoli. Some SH2B1δ is also present in the plasma membrane and nucleus. Nucleolar localization, driven by two highly basic regions unique to SH2B1δ, is required for SH2B1δ to maximally increase neurite complexity and BDNF-induced expression of Egr1, Arc and FosL1.
Collapse
Affiliation(s)
- Jessica L. Cote
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul B. Vander
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael Ellis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joel M. Cline
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nadezhda Svezhova
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael E. Doche
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Travis J. Maures
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tahrim A. Choudhury
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Seongbae Kong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Olivia G. J. Klaft
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ray M. Joe
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lawrence S. Argetsinger
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christin Carter-Su
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Kim CH, Kim S, Kim SH, Roh J, Jin H, Song B. Role of densin-180 in mouse ventral hippocampal neurons in 24-hr retention of contextual fear conditioning. Brain Behav 2020; 10:e01891. [PMID: 33064361 PMCID: PMC7749528 DOI: 10.1002/brb3.1891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/01/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Densin-180 interacts with postsynaptic molecules including calcium/calmodulin-dependent protein kinase IIα (CaMKIIα) but its function in learning and memory process has been unclear. METHODS To investigate a role of hippocampal densin-180 in contextual fear conditioning (CFC) learning and memory processes, knockdown (KD) of densin-180 in hippocampal subareas was applied. RESULTS First, ventral hippocampal (vHC) densin-180 KD impaired single-trial CFC (stCFC) memory one day later. stCFC caused freezing behaviors to reach the peak about one hour later in both control and KD mice, but then freezing was disappeared at 2 hr postshock in KD mice. Second, stCFC caused an immediate and transient reduction of vHC densin-180 in control mice, which was not observed in KD mice. Third, stCFC caused phosphorylated-T286 (p-T286) CaMKIIα to change similarly to densin-180, but p-T305 CaMKIIα was increased 1 hr later in control mice. In KD mice, these effects were gone. Moreover, both basal levels of p-T286 and p-T305 CaMKIIα were reduced without change in total CaMKIIα in KD mice. Fourth, we found double-trial CFC (dtCFC) memory acquisition and retrieval kinetics were different from those of stCFC in vHC KD mice. In addition, densin-180 in dorsal hippocampal area appeared to play its unique role during the very early retrieval period of both CFC memories. CONCLUSION This study shows that vHC densin-180 is necessary for stCFC memory formation and retrieval and suggests that both densin-180 and p-T305 CaMKIIα at 1 ~ 2 hr postshock are important for stCFC memory formation. We conclude that roles of hippocampal neuronal densin-180 in CFC are temporally dynamic and differential depending on the pattern of conditioning stimuli and its location along the dorsoventral axis of hippocampal formation.
Collapse
Affiliation(s)
- Chong-Hyun Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea.,Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Seoyul Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea.,Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Su-Hyun Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea.,Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Jongtae Roh
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea.,Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Harin Jin
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea.,Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Bokyung Song
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea.,Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| |
Collapse
|
8
|
Brancato A, Castelli V, Lavanco G, Marino RAM, Cannizzaro C. In utero Δ9-tetrahydrocannabinol exposure confers vulnerability towards cognitive impairments and alcohol drinking in the adolescent offspring: Is there a role for neuropeptide Y? J Psychopharmacol 2020; 34:663-679. [PMID: 32338122 DOI: 10.1177/0269881120916135] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cannabinoid consumption during pregnancy has been increasing on the wave of the broad-based legalisation of cannabis in Western countries, raising concern about the putative detrimental outcomes on foetal neurodevelopment. Indeed, since the endocannabinoid system regulates synaptic plasticity, emotional and cognitive processes from early stages of life interfering with it and other excitability endogenous modulators, such as neuropeptide Y (NPY), might contribute to the occurrence of a vulnerable phenotype later in life. AIMS This research investigated whether in utero exposure to Δ9-tetrahydrocannabinol (THC) may induce deficits in emotional/cognitive processes and alcohol vulnerability in adolescent offspring. NPY and excitatory postsynaptic density (PSD) machinery were measured as markers of neurobiological vulnerability. METHODS Following in utero THC exposure (2 mg/kg delivered subcutaneously), preadolescent male rat offspring were assessed for: behavioural reactivity in the open field test, neutral declarative memory and aversive limbic memory in the Novel Object and Emotional Object Recognition tests, immunofluorescence for NPY neurons and the PSD proteins Homer-1, 1b/c and 2 in the prefrontal cortex, amygdala and nucleus accumbens at adolescence (cohort 1); and instrumental learning, alcohol taking, relapse and conflict behaviour in the operant chamber throughout adolescence until early adulthood (cohort 2). RESULTS In utero THC-exposed adolescent rats showed: (a) increased locomotor activity; (b) no alteration in neutral declarative memory; (c) impaired aversive limbic memory; (d) decreased NPY-positive neurons in limbic regions; (e) region-specific variations in Homer-1, 1b/c and 2 immunoreactivity; (f) decreased instrumental learning and increased alcohol drinking, relapse and conflict behaviour in the operant chamber. CONCLUSION Gestational THC impaired the formation of memory traces when integration between environmental encoding and emotional/motivational processing was required and promoted the development of alcohol-addictive behaviours. The abnormalities in NPY signalling and PSD make-up may represent the common neurobiological background, suggesting new targets for future research.
Collapse
Affiliation(s)
- Anna Brancato
- Department of Health Promotion, Mother-Child Care, Internal Medicine and Medical Specialties of Excellence 'G. D'Alessandro', University of Palermo, Palermo, Italy
| | - Valentina Castelli
- Department of Health Promotion, Mother-Child Care, Internal Medicine and Medical Specialties of Excellence 'G. D'Alessandro', University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Gianluca Lavanco
- INSERM U1215, NeuroCentre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France.,Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Rosa Anna Maria Marino
- Department of Anatomy and Neurobiology, School of Medicine, University of Maryland, Baltimore, USA
| | - Carla Cannizzaro
- Department of Health Promotion, Mother-Child Care, Internal Medicine and Medical Specialties of Excellence 'G. D'Alessandro', University of Palermo, Palermo, Italy
| |
Collapse
|
9
|
Ahamad A, Wang J, Ge S, Kirschen GW. Early Dendritic Morphogenesis of Adult-Born Dentate Granule Cells Is Regulated by FHL2. Front Neurosci 2020; 14:202. [PMID: 32256309 PMCID: PMC7090230 DOI: 10.3389/fnins.2020.00202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022] Open
Abstract
Dentate granule cells (DGCs), the progeny of neural stem cells (NSCs) in the sub-granular zone of the dentate gyrus (DG), must develop and functionally integrate with the mature cohort of neurons in order to maintain critical hippocampal functions throughout adulthood. Dysregulation in the continuum of DGC development can result in aberrant morphology and disrupted functional maturation, impairing neuroplasticity of the network. Yet, the molecular underpinnings of the signaling involved in adult-born DGC maturation including dendritic growth, which correlates with functional integration, remains incompletely understood. Given the high metabolic activity in the dentate gyrus (DG) required to achieve continuous neurogenesis, we investigated the potential regulatory role of a cellular metabolism-linked gene recently implicated in NSC cycling and neuroblast migration, called Four and a half LIM domain 2 (FHL2). The FHL2 protein modulates numerous pathways related to proliferation, migration, survival and cytoskeletal rearrangement in peripheral tissues, interacting with the machinery of the sphingosine-1-phosphate pathway, also known to be highly active especially in the hippocampus. Yet, the potential relevance of FHL2 to adult-born DGC development remains unknown. To elucidate the role of FHL2 in DGC development in the adult brain, we first confirmed the endogenous expression of FHL2 in NSCs and new granule cells within the DG, then engineered viral vectors for genetic manipulation experiments, investigating morphological changes in early stages of DGC development. Overexpression of FHL2 during early DGC development resulted in marked sprouting and branching of dendrites, while silencing of FHL2 increased dendritic length. Together, these findings suggest a novel role of FHL2 in adult-born DGC morphological maturation, which may open up a new line of investigation regarding the relevance of this gene in physiology and pathologies of the hippocampus such as mesial temporal lobe epilepsy (MTLE).
Collapse
Affiliation(s)
- Afrinash Ahamad
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, United States.,School of Health Technology and Management, Stony Brook University, Stony Brook, NY, United States
| | - Jia Wang
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
| | - Gregory W Kirschen
- Medical Scientist Training Program (MSTP), Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
10
|
Amacher JF, Brooks L, Hampton TH, Madden DR. Specificity in PDZ-peptide interaction networks: Computational analysis and review. JOURNAL OF STRUCTURAL BIOLOGY-X 2020; 4:100022. [PMID: 32289118 PMCID: PMC7138185 DOI: 10.1016/j.yjsbx.2020.100022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 01/03/2023]
Abstract
Globular PDZ domains typically serve as protein-protein interaction modules that regulate a wide variety of cellular functions via recognition of short linear motifs (SLiMs). Often, PDZ mediated-interactions are essential components of macromolecular complexes, and disruption affects the entire scaffold. Due to their roles as linchpins in trafficking and signaling pathways, PDZ domains are attractive targets: both for controlling viral pathogens, which bind PDZ domains and hijack cellular machinery, as well as for developing therapies to combat human disease. However, successful therapeutic interventions that avoid off-target effects are a challenge, because each PDZ domain interacts with a number of cellular targets, and specific binding preferences can be difficult to decipher. Over twenty-five years of research has produced a wealth of data on the stereochemical preferences of individual PDZ proteins and their binding partners. Currently the field lacks a central repository for this information. Here, we provide this important resource and provide a manually curated, comprehensive list of the 271 human PDZ domains. We use individual domain, as well as recent genomic and proteomic, data in order to gain a holistic view of PDZ domains and interaction networks, arguing this knowledge is critical to optimize targeting selectivity and to benefit human health.
Collapse
Affiliation(s)
- Jeanine F Amacher
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Department of Chemistry, Western Washington University, Bellingham, WA 98225, USA
| | - Lionel Brooks
- Department of Biology, Western Washington University, Bellingham, WA 98225, USA
| | - Thomas H Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Dean R Madden
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
11
|
SHANK2 mutations associated with autism spectrum disorder cause hyperconnectivity of human neurons. Nat Neurosci 2019; 22:556-564. [PMID: 30911184 PMCID: PMC6475597 DOI: 10.1038/s41593-019-0365-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Heterozygous loss-of-function mutations in SHANK2 are associated with autism spectrum disorder (ASD). We generated cortical neurons from induced pluripotent stem cells (iPSC) derived from neurotypic and ASD-affected donors. We developed Sparse coculture for Connectivity (SparCon) assays where SHANK2 and control neurons were differentially labeled and sparsely seeded together on a lawn of unlabeled control neurons. We observed increases in dendrite length, dendrite complexity, synapse number, and frequency of spontaneous excitatory postsynaptic currents. These findings were phenocopied in gene-edited homozygous SHANK2 knockout cells and rescued by gene correction of an ASD SHANK2 mutation. Dendrite length increases were exacerbated by IGF1, TG003, or BDNF, and suppressed by DHPG treatment. The transcriptome in isogenic SHANK2 neurons was perturbed in synapse, plasticity, and neuronal morphogenesis gene sets and ASD gene modules, and activity-dependent dendrite extension was impaired. Our findings provide evidence for hyperconnectivity and altered transcriptome in SHANK2 neurons derived from ASD subjects.
Collapse
|
12
|
Zhang-James Y, Fernàndez-Castillo N, Hess JL, Malki K, Glatt SJ, Cormand B, Faraone SV. An integrated analysis of genes and functional pathways for aggression in human and rodent models. Mol Psychiatry 2019; 24:1655-1667. [PMID: 29858598 PMCID: PMC6274606 DOI: 10.1038/s41380-018-0068-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/04/2018] [Accepted: 04/03/2018] [Indexed: 11/12/2022]
Abstract
Human genome-wide association studies (GWAS), transcriptome analyses of animal models, and candidate gene studies have advanced our understanding of the genetic architecture of aggressive behaviors. However, each of these methods presents unique limitations. To generate a more confident and comprehensive view of the complex genetics underlying aggression, we undertook an integrated, cross-species approach. We focused on human and rodent models to derive eight gene lists from three main categories of genetic evidence: two sets of genes identified in GWAS studies, four sets implicated by transcriptome-wide studies of rodent models, and two sets of genes with causal evidence from online Mendelian inheritance in man (OMIM) and knockout (KO) mice reports. These gene sets were evaluated for overlap and pathway enrichment to extract their similarities and differences. We identified enriched common pathways such as the G-protein coupled receptor (GPCR) signaling pathway, axon guidance, reelin signaling in neurons, and ERK/MAPK signaling. Also, individual genes were ranked based on their cumulative weights to quantify their importance as risk factors for aggressive behavior, which resulted in 40 top-ranked and highly interconnected genes. The results of our cross-species and integrated approach provide insights into the genetic etiology of aggression.
Collapse
Affiliation(s)
- Yanli Zhang-James
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY, USA.
| | - Noèlia Fernàndez-Castillo
- 0000 0004 1937 0247grid.5841.8Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain ,0000 0004 1791 1185grid.452372.5Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain ,0000 0004 1937 0247grid.5841.8Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain ,Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Spain
| | - Jonathan L Hess
- 0000 0000 9159 4457grid.411023.5Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY USA
| | - Karim Malki
- 0000 0001 2322 6764grid.13097.3cKing’s College London, MRC Social, Genetic and Developmental Psychiatry Centre at the Institute of Psychiatry, Psychology and Neuroscience (IOPPN), London, UK
| | - Stephen J Glatt
- 0000 0000 9159 4457grid.411023.5Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY USA ,0000 0000 9159 4457grid.411023.5Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, NY USA
| | - Bru Cormand
- 0000 0004 1937 0247grid.5841.8Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain ,0000 0004 1791 1185grid.452372.5Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain ,0000 0004 1937 0247grid.5841.8Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain ,Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Spain
| | - Stephen V Faraone
- 0000 0000 9159 4457grid.411023.5Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, NY USA ,0000 0000 9159 4457grid.411023.5Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, NY USA ,0000 0004 1936 7443grid.7914.bK.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| |
Collapse
|
13
|
Bhattacharya D, Majrashi M, Ramesh S, Govindarajulu M, Bloemer J, Fujihashi A, Crump BR, Hightower H, Bhattacharya S, Moore T, Suppiramaniam V, Dhanasekaran M. Assessment of the cerebellar neurotoxic effects of nicotine in prenatal alcohol exposure in rats. Life Sci 2017; 194:177-184. [PMID: 29225110 DOI: 10.1016/j.lfs.2017.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/27/2017] [Accepted: 12/06/2017] [Indexed: 01/12/2023]
Abstract
The adverse effects of prenatal nicotine and alcohol exposure on human reproductive outcomes are a major scientific and public health concern. In the United States, substantial percentage of women (20-25%) of childbearing age currently smoke cigarettes and consume alcohol, and only a small percentage of these individuals quit after learning of their pregnancy. However, there are very few scientific reports on the effect of nicotine in prenatal alcohol exposure on the cerebellum of the offspring. Therefore, this study was conducted to investigate the cerebellar neurotoxic effects of nicotine in a rodent model of Fetal Alcohol Spectrum Disorder (FASD). In this study, we evaluated the behavioral changes, biochemical markers of oxidative stress and apoptosis, mitochondrial functions and the molecular mechanisms associated with nicotine in prenatal alcohol exposure on the cerebellum. Prenatal nicotine and alcohol exposure induced oxidative stress, did not affect the mitochondrial functions, increased the monoamine oxidase activity, increased caspase expression and decreased ILK, PSD-95 and GLUR1 expression without affecting the GSK-3β. Thus, our current study of prenatal alcohol and nicotine exposure on cerebellar neurotoxicity may lead to new scientific perceptions and novel and suitable therapeutic actions in the future.
Collapse
Affiliation(s)
| | - Mohammed Majrashi
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Sindhu Ramesh
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Manoj Govindarajulu
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Jenna Bloemer
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Ayaka Fujihashi
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Bailee-Ryan Crump
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Harrison Hightower
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | | | - Timothy Moore
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Vishnu Suppiramaniam
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | | |
Collapse
|
14
|
Tomasetti C, Iasevoli F, Buonaguro EF, De Berardis D, Fornaro M, Fiengo ALC, Martinotti G, Orsolini L, Valchera A, Di Giannantonio M, de Bartolomeis A. Treating the Synapse in Major Psychiatric Disorders: The Role of Postsynaptic Density Network in Dopamine-Glutamate Interplay and Psychopharmacologic Drugs Molecular Actions. Int J Mol Sci 2017; 18:E135. [PMID: 28085108 PMCID: PMC5297768 DOI: 10.3390/ijms18010135] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/25/2016] [Accepted: 01/08/2017] [Indexed: 12/20/2022] Open
Abstract
Dopamine-glutamate interplay dysfunctions have been suggested as pathophysiological key determinants of major psychotic disorders, above all schizophrenia and mood disorders. For the most part, synaptic interactions between dopamine and glutamate signaling pathways take part in the postsynaptic density, a specialized ultrastructure localized under the membrane of glutamatergic excitatory synapses. Multiple proteins, with the role of adaptors, regulators, effectors, and scaffolds compose the postsynaptic density network. They form structural and functional crossroads where multiple signals, starting at membrane receptors, are received, elaborated, integrated, and routed to appropriate nuclear targets. Moreover, transductional pathways belonging to different receptors may be functionally interconnected through postsynaptic density molecules. Several studies have demonstrated that psychopharmacologic drugs may differentially affect the expression and function of postsynaptic genes and proteins, depending upon the peculiar receptor profile of each compound. Thus, through postsynaptic network modulation, these drugs may induce dopamine-glutamate synaptic remodeling, which is at the basis of their long-term physiologic effects. In this review, we will discuss the role of postsynaptic proteins in dopamine-glutamate signals integration, as well as the peculiar impact of different psychotropic drugs used in clinical practice on postsynaptic remodeling, thereby trying to point out the possible future molecular targets of "synapse-based" psychiatric therapeutic strategies.
Collapse
Affiliation(s)
- Carmine Tomasetti
- NHS, Department of Mental Health ASL Teramo, Psychiatric Service of Diagnosis and Treatment, Hospital "Maria SS dello Splendore", 641021 Giulianova, Italy.
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
- Polyedra Research Group, 64100 Teramo, Italy.
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
- Polyedra Research Group, 64100 Teramo, Italy.
| | - Elisabetta Filomena Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
- Polyedra Research Group, 64100 Teramo, Italy.
| | - Domenico De Berardis
- Polyedra Research Group, 64100 Teramo, Italy.
- NHS, Department of Mental Health ASL Teramo, Psychiatric Service of Diagnosis and Treatment, Hospital "G. Mazzini", 64100 Teramo, Italy.
- Department of Neuroscience and Imaging, University "G. d'Annunzio", 66100 Chieti, Italy.
| | - Michele Fornaro
- Polyedra Research Group, 64100 Teramo, Italy.
- New York State Psychiatric Institute, Columbia University, New York, NY 10027, USA.
| | | | - Giovanni Martinotti
- Polyedra Research Group, 64100 Teramo, Italy.
- Department of Neuroscience and Imaging, University "G. d'Annunzio", 66100 Chieti, Italy.
| | - Laura Orsolini
- Polyedra Research Group, 64100 Teramo, Italy.
- Casa di Cura Villa San Giuseppe, 63100 Ascoli Piceno, Italy.
| | - Alessandro Valchera
- Polyedra Research Group, 64100 Teramo, Italy.
- Casa di Cura Villa San Giuseppe, 63100 Ascoli Piceno, Italy.
| | | | - Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
| |
Collapse
|
15
|
Zhang LM, Wang YL, Liu YQ, Xue R, Zhang YZ, Yang RF, Li YF. Antidepressant-like effects of YL-IPA08, a potent ligand for the translocator protein (18 kDa) in chronically stressed rats. Neuropharmacology 2016; 113:567-575. [PMID: 27845056 DOI: 10.1016/j.neuropharm.2016.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 10/19/2016] [Accepted: 11/02/2016] [Indexed: 10/20/2022]
Abstract
The present study aimed to examine the molecular and cellular mechanisms underlying the antidepressant-like effect of YL-IPA08, a novel TSPO ligand designed and synthesized at our institute. We firstly used the chronic unpredictable stress (CUS) procedure of rats, a well validated stress-related animal model of depression, to further determine the antidepressant-like of YL-IPA08. And we found that YL-IPA08 caused significant suppression of inhibiting of locomotor activity, reducing the sucrose preference and increasing the latency to eat induced by CUS. In addition, YL-IPA08 treatment increased the levels of progesterone and allopregnanolone in the hippocampus and prefrontal cortex of post- CUS rats. Furthermore, long-term YL-IPA08 administration reversed dendritic shrinkage, down-regulation of neurotrophic signaling pathway within the hippocampus, as well as HPA dysfunctions simultaneously observed in the CUS rats. Collectively, the evidence presented above supports the notion that binding to TSPO and the subsequent synthesis of neurosteroid, maintenance of hippocampal morphologic and functional plasticity, and preventing HPA axis dysfunction, may account for the profound molecular and cellular mechanism underlying the antidepressant-like effect of YL-IPA08.
Collapse
Affiliation(s)
- Li-Ming Zhang
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yu-Lu Wang
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Yan-Qin Liu
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Rui Xue
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - You-Zhi Zhang
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ri-Fang Yang
- Department of Medicinal Chemistry, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Yun-Feng Li
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
16
|
Geiger JC, Lipka J, Segura I, Hoyer S, Schlager MA, Wulf PS, Weinges S, Demmers J, Hoogenraad CC, Acker-Palmer A. The GRIP1/14-3-3 pathway coordinates cargo trafficking and dendrite development. Dev Cell 2014; 28:381-93. [PMID: 24576423 DOI: 10.1016/j.devcel.2014.01.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/16/2014] [Accepted: 01/21/2014] [Indexed: 01/22/2023]
Abstract
Regulation of cargo transport via adaptor molecules is essential for neuronal development. However, the role of PDZ scaffolding proteins as adaptors in neuronal cargo trafficking is still poorly understood. Here, we show by genetic deletion in mice that the multi-PDZ domain scaffolding protein glutamate receptor interacting protein 1 (GRIP1) is required for dendrite development. We identify an interaction between GRIP1 and 14-3-3 proteins that is essential for the function of GRIP1 as an adaptor protein in dendritic cargo transport. Mechanistically, 14-3-3 binds to the kinesin-1 binding region in GRIP1 in a phospho-dependent manner and detaches GRIP1 from the kinesin-1 motor protein complex thereby regulating cargo transport. A single point mutation in the Thr956 of GRIP1 in transgenic mice impairs dendritic development. Together, our results show a regulatory role for GRIP1 during microtubule-based transport and suggest a crucial function for 14-3-3 proteins in controlling kinesin-1 motor attachment during neuronal development.
Collapse
Affiliation(s)
- Julia C Geiger
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60438 Frankfurt am Main, Germany; Focus Program Translational Neurosciences (FTN), University of Mainz, 55131 Mainz, Germany
| | - Joanna Lipka
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH Utrecht, the Netherlands; International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Inmaculada Segura
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60438 Frankfurt am Main, Germany
| | - Susanne Hoyer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60438 Frankfurt am Main, Germany
| | - Max A Schlager
- Department of Neuroscience, Erasmus Medical Center, 3015GE Rotterdam, the Netherlands
| | - Phebe S Wulf
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH Utrecht, the Netherlands; Department of Neuroscience, Erasmus Medical Center, 3015GE Rotterdam, the Netherlands
| | - Stefan Weinges
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60438 Frankfurt am Main, Germany
| | - Jeroen Demmers
- Proteomics Center, Erasmus Medical Center, 3015GE Rotterdam, the Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584CH Utrecht, the Netherlands; Department of Neuroscience, Erasmus Medical Center, 3015GE Rotterdam, the Netherlands.
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60438 Frankfurt am Main, Germany; Focus Program Translational Neurosciences (FTN), University of Mainz, 55131 Mainz, Germany.
| |
Collapse
|
17
|
Wippel C, Maurer J, Förtsch C, Hupp S, Bohl A, Ma J, Mitchell TJ, Bunkowski S, Brück W, Nau R, Iliev AI. Bacterial cytolysin during meningitis disrupts the regulation of glutamate in the brain, leading to synaptic damage. PLoS Pathog 2013; 9:e1003380. [PMID: 23785278 PMCID: PMC3681734 DOI: 10.1371/journal.ppat.1003380] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 04/08/2013] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae (pneumococcal) meningitis is a common bacterial infection of the brain. The cholesterol-dependent cytolysin pneumolysin represents a key factor, determining the neuropathogenic potential of the pneumococci. Here, we demonstrate selective synaptic loss within the superficial layers of the frontal neocortex of post-mortem brain samples from individuals with pneumococcal meningitis. A similar effect was observed in mice with pneumococcal meningitis only when the bacteria expressed the pore-forming cholesterol-dependent cytolysin pneumolysin. Exposure of acute mouse brain slices to only pore-competent pneumolysin at disease-relevant, non-lytic concentrations caused permanent dendritic swelling, dendritic spine elimination and synaptic loss. The NMDA glutamate receptor antagonists MK801 and D-AP5 reduced this pathology. Pneumolysin increased glutamate levels within the mouse brain slices. In mouse astrocytes, pneumolysin initiated the release of glutamate in a calcium-dependent manner. We propose that pneumolysin plays a significant synapto- and dendritotoxic role in pneumococcal meningitis by initiating glutamate release from astrocytes, leading to subsequent glutamate-dependent synaptic damage. We outline for the first time the occurrence of synaptic pathology in pneumococcal meningitis and demonstrate that a bacterial cytolysin can dysregulate the control of glutamate in the brain, inducing excitotoxic damage.
Collapse
Affiliation(s)
- Carolin Wippel
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Jana Maurer
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Christina Förtsch
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Sabrina Hupp
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Alexandra Bohl
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| | - Jiangtao Ma
- Division of Infection and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Timothy J. Mitchell
- Division of Infection and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, United Kingdom
- Chair of Microbial Infection and Immunity, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stephanie Bunkowski
- Department of Neuropathology, Georg-August-University of Göttingen, Göttingen, Germany
| | - Wolfgang Brück
- Department of Neuropathology, Georg-August-University of Göttingen, Göttingen, Germany
| | - Roland Nau
- Department of Neuropathology, Georg-August-University of Göttingen, Göttingen, Germany
- Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Asparouh I. Iliev
- DFG Membrane/Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
18
|
Liu DC, Jow GM, Chuang CC, Peng YJ, Hsu PH, Tang CY. Densin-180 is Not a Transmembrane Protein. Cell Biochem Biophys 2013; 67:773-83. [PMID: 23516094 DOI: 10.1007/s12013-013-9570-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Dai-Chi Liu
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
19
|
High dose zinc supplementation induces hippocampal zinc deficiency and memory impairment with inhibition of BDNF signaling. PLoS One 2013; 8:e55384. [PMID: 23383172 PMCID: PMC3561272 DOI: 10.1371/journal.pone.0055384] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/21/2012] [Indexed: 11/21/2022] Open
Abstract
Zinc ions highly concentrate in hippocampus and play a key role in modulating spatial learning and memory. At a time when dietary fortification and supplementation of zinc have increased the zinc consuming level especially in the youth, the toxicity of zinc overdose on brain function was underestimated. In the present study, weaning ICR mice were given water supplemented with 15 ppm Zn (low dose), 60 ppm Zn (high dose) or normal lab water for 3 months, the behavior and brain zinc homeostasis were tested. Mice fed high dose of zinc showed hippocampus-dependent memory impairment. Unexpectedly, zinc deficiency, but not zinc overload was observed in hippocampus, especially in the mossy fiber-CA3 pyramid synapse. The expression levels of learning and memory related receptors and synaptic proteins such as NMDA-NR2A, NR2B, AMPA-GluR1, PSD-93 and PSD-95 were significantly decreased in hippocampus, with significant loss of dendritic spines. In keeping with these findings, high dose intake of zinc resulted in decreased hippocampal BDNF level and TrkB neurotrophic signaling. At last, increasing the brain zinc level directly by brain zinc injection induced BDNF expression, which was reversed by zinc chelating in vivo. These results indicate that zinc plays an important role in hippocampus-dependent learning and memory and BDNF expression, high dose supplementation of zinc induces specific zinc deficiency in hippocampus, which further impair learning and memory due to decreased availability of synaptic zinc and BDNF deficit.
Collapse
|
20
|
Paschou M, Paraskevopoulou MD, Vlachos IS, Koukouraki P, Hatzigeorgiou AG, Doxakis E. miRNA regulons associated with synaptic function. PLoS One 2012; 7:e46189. [PMID: 23071543 PMCID: PMC3468272 DOI: 10.1371/journal.pone.0046189] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 08/28/2012] [Indexed: 11/19/2022] Open
Abstract
Differential RNA localization and local protein synthesis regulate synapse function and plasticity in neurons. MicroRNAs are a conserved class of regulatory RNAs that control mRNA stability and translation in tissues. They are abundant in the brain but the extent into which they are involved in synaptic mRNA regulation is poorly known. Herein, a computational analysis of the coding and 3′UTR regions of 242 presynaptic and 304 postsynaptic proteins revealed that 91% of them are predicted to be microRNA targets. Analysis of the longest 3′UTR isoform of synaptic transcripts showed that presynaptic mRNAs have significantly longer 3′UTR than control and postsynaptic mRNAs. In contrast, the shortest 3′UTR isoform of postsynaptic mRNAs is significantly shorter than control and presynaptic mRNAs, indicating they avert microRNA regulation under specific conditions. Examination of microRNA binding site density of synaptic 3′UTRs revealed that they are twice as dense as the rest of protein-coding transcripts and that approximately 50% of synaptic transcripts are predicted to have more than five different microRNA sites. An interaction map exploring the association of microRNAs and their targets revealed that a small set of ten microRNAs is predicted to regulate 77% and 80% of presynaptic and postsynaptic transcripts, respectively. Intriguingly, many of these microRNAs have yet to be identified outside primate mammals, implicating them in cognition differences observed between high-level primates and non-primate mammals. Importantly, the identified miRNAs have been previously associated with psychotic disorders that are characterized by neural circuitry dysfunction, such as schizophrenia. Finally, molecular dissection of their KEGG pathways showed enrichment for neuronal and synaptic processes. Adding on current knowledge, this investigation revealed the extent of miRNA regulation at the synapse and predicted critical microRNAs that would aid future research on the control of neuronal plasticity and etiology of psychiatric diseases.
Collapse
Affiliation(s)
- Maria Paschou
- Basic Neurosciences Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria D. Paraskevopoulou
- Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming” Vari, Greece
| | - Ioannis S. Vlachos
- Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming” Vari, Greece
| | - Pelagia Koukouraki
- Basic Neurosciences Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Artemis G. Hatzigeorgiou
- Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming” Vari, Greece
- Department of Computer and Communication Engineering, University of Thessaly, Volos, Greece
| | - Epaminondas Doxakis
- Basic Neurosciences Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
21
|
Cohen S, Kozlovsky N, Matar MA, Kaplan Z, Zohar J, Cohen H. Post-exposure sleep deprivation facilitates correctly timed interactions between glucocorticoid and adrenergic systems, which attenuate traumatic stress responses. Neuropsychopharmacology 2012; 37:2388-404. [PMID: 22713910 PMCID: PMC3442354 DOI: 10.1038/npp.2012.94] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Reliable evidence supports the role of sleep in learning and memory processes. In rodents, sleep deprivation (SD) negatively affects consolidation of hippocampus-dependent memories. As memory is integral to post-traumatic stress symptoms, the effects of post-exposure SD on various aspect of the response to stress in a controlled, prospective animal model of post-traumatic stress disorder (PTSD) were evaluated. Rats were deprived of sleep for 6 h throughout the first resting phase after predator scent stress exposure. Behaviors in the elevated plus-maze and acoustic startle response tests were assessed 7 days later, and served for classification into behavioral response groups. Freezing response to a trauma reminder was assessed on day 8. Urine samples were collected daily for corticosterone levels, and heart rate (HR) was also measured. Finally, the impact of manipulating the hypothalamus-pituitary-adrenal axis and adrenergic activity before SD was assessed. Mifepristone (MIFE) and epinephrine (EPI) were administered systemically 10-min post-stress exposure and behavioral responses and response to trauma reminder were measured on days 7-8. Hippocampal expression of glucocorticoid receptors (GRs) and morphological assessment of arborization and dendritic spines were subsequently evaluated. Post-exposure SD effectively ameliorated long-term, stress-induced, PTSD-like behavioral disruptions, reduced trauma reminder freezing responses, and decreased hippocampal expression of GR compared with exposed-untreated controls. Although urine corticosterone levels were significantly elevated 1 h after SD and the HR was attenuated, antagonizing GRs with MIFE or stimulation of adrenergic activity with EPI effectively abolished the effect of SD. MIFE- and EPI-treated animals clearly demonstrated significantly lower total dendritic length, fewer branches and lower spine density along dentate gyrus dendrites with increased levels of GR expression 8 days after exposure, as compared with exposed-SD animals. Intentional prevention of sleep in the early aftermath of stress exposure may well be beneficial in attenuating traumatic stress-related sequelae. Post-exposure SD may disrupt the consolidation of aversive or fearful memories by facilitating correctly timed interactions between glucocorticoid and adrenergic systems.
Collapse
Affiliation(s)
- Shlomi Cohen
- Department of Psychology, Ben-Gurion University of the Negev, Beer Sheva, Israel,Anxiety and Stress Research Unit, Ministry of Health Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Nitsan Kozlovsky
- Anxiety and Stress Research Unit, Ministry of Health Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael A Matar
- Anxiety and Stress Research Unit, Ministry of Health Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Zeev Kaplan
- Anxiety and Stress Research Unit, Ministry of Health Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Joseph Zohar
- Division of Psychiatry, Department of Psychiatry, The Chaim Sheba Medical Center, Sackler Medical School, Tel-Aviv University, Tel-Hashomer, Israel
| | - Hagit Cohen
- Department of Psychology, Ben-Gurion University of the Negev, Beer Sheva, Israel,Anxiety and Stress Research Unit, Ministry of Health Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel,Anxiety and Stress Research Unit, Ministry of Health Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, PO Box 4600, Beer-Sheva 84170, Israel, Tel: +972 8 6401743, Fax: +972 8 6401742, E-mail:
| |
Collapse
|
22
|
MacDonald JIS, Dietrich A, Gamble S, Hryciw T, Grant RI, Meakin SO. Nesca, a novel neuronal adapter protein, links the molecular motor kinesin with the pre-synaptic membrane protein, syntaxin-1, in hippocampal neurons. J Neurochem 2012; 121:861-80. [PMID: 22404429 DOI: 10.1111/j.1471-4159.2012.07729.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Vesicular transport in neurons plays a vital role in neuronal function and survival. Nesca is a novel protein that we previously identified and herein describe its pattern of expression, subcellular localization and protein-protein interactions both in vitro and in vivo. Specifically, a large proportion of Nesca is in tight association with both actin and microtubule cytoskeletal proteins. Nesca binds to F-actin, microtubules, βIII and acetylated α-tubulin, but not neurofilaments or the actin-binding protein drebrin, in in vitro-binding assays. Nesca co-immunoprecipitates with kinesin heavy chain (KIF5B) and kinesin light-chain motors as well as with the synaptic membrane precursor protein, syntaxin-1, and is a constituent of the post-synaptic density. Moreover, in vitro-binding assays indicate that Nesca directly binds KIF5B, kinesin light-chain and syntaxin-1. In contrast, Nesca does not co-immunoprecipitate with the kinesin motors KIF1B, KIF3A nor does it bind syntaxin-4 or the synaptosome-associated protein 25 kDa (SNAP-25) in vitro. Nesca expression in neurons is highly punctuate, co-stains with syntaxin-1, and is found in fractions containing markers of early endosomes and Golgi suggesting that it is involved in vesicular transport. Collectively, these data suggest that Nesca functions as an adapter involved in neuronal vesicular transport including vesicles containing soluble N-ethylmaleimide sensitive factor attachment protein receptors that are essential to exocytosis.
Collapse
Affiliation(s)
- James I S MacDonald
- Laboratory of Neural Signaling, Molecular Brain Research Group, Robarts Research Institute, Schulich School of Medicine, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | |
Collapse
|
23
|
Zohar J, Yahalom H, Kozlovsky N, Cwikel-Hamzany S, Matar MA, Kaplan Z, Yehuda R, Cohen H. High dose hydrocortisone immediately after trauma may alter the trajectory of PTSD: interplay between clinical and animal studies. Eur Neuropsychopharmacol 2011; 21:796-809. [PMID: 21741804 DOI: 10.1016/j.euroneuro.2011.06.001] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 05/12/2011] [Accepted: 06/05/2011] [Indexed: 02/06/2023]
Abstract
High-dose corticosteroids have been reported to reduce symptoms of acute stress and post-traumatic stress in polytrauma patients and in animal studies. The underlying mechanism of action remains largely unclear. These issues were addressed in parallel in the clinical and preclinical studies below. In this preliminary study, 25 patients with acute stress symptoms were administered a single intravenous bolus of high-dose hydrocortisone (100-140 mg) or placebo within 6 h of a traumatic event in a prospective, randomized, double-blind, placebo-controlled pilot study. Early single high-dose hydrocortisone intervention attenuated the core symptoms of both the acute stress and of subsequent PTSD in patients. High-dose hydrocortisone treatment given in the first few hours after a traumatic experience was associated with significant favorable changes in the trajectory of exposure to trauma, as expressed by the reduced risk of the development of PTSD post-trauma. In parallel, a comparative study of morphological arborization in dentate gyrus and its modulating molecules was performed in stress-exposed animals treated with high-dose hydrocortisone. Steroid-treated stressed animals displayed significantly increased dendritic growth and spine density, with increased levels of brain-derived neurotrophic factor (BDNF) and obtunded postsynaptic density-95 (PSD-95) levels. The animal study provided insights into the potential mechanism of this intervention, as it identified relevant morphological and biochemical associations to the clinical observations. Thus, evidence from clinical and animal studies suggests that there is a "window of opportunity" in the early aftermath of trauma to help those who are vulnerable to the development of chronic PTSD.
Collapse
Affiliation(s)
- Joseph Zohar
- Division of Psychiatry, The State of Israel Ministry of Health, The Chaim Sheba Medical Center, Sackler Medical School, Tel-Aviv University, Tel Hashomer, Israel
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Yasuda Y, Hashimoto R, Yamamori H, Ohi K, Fukumoto M, Umeda-Yano S, Mohri I, Ito A, Taniike M, Takeda M. Gene expression analysis in lymphoblasts derived from patients with autism spectrum disorder. Mol Autism 2011; 2:9. [PMID: 21615902 PMCID: PMC3118341 DOI: 10.1186/2040-2392-2-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 05/26/2011] [Indexed: 12/22/2022] Open
Abstract
Background The autism spectrum disorders (ASDs) are complex neurodevelopmental disorders that result in severe and pervasive impairment in the development of reciprocal social interaction and verbal and nonverbal communication skills. In addition, individuals with ASD have stereotypical behavior, interests and activities. Rare mutations of some genes, such as neuroligin (NLGN) 3/4, neurexin (NRXN) 1, SHANK3, MeCP2 and NHE9, have been reported to be associated with ASD. In the present study, we investigated whether alterations in mRNA expression levels of these genes could be found in lymphoblastoid cell lines derived from patients with ASD. Methods We measured mRNA expression levels of NLGN3/4, NRXN1, SHANK3, MeCP2, NHE9 and AKT1 in lymphoblastoid cells from 35 patients with ASD and 35 healthy controls, as well as from 45 patients with schizophrenia and 45 healthy controls, using real-time quantitative reverse transcriptase polymerase chain reaction assays. Results The mRNA expression levels of NLGN3 and SHANK3 normalized by β-actin or TBP were significantly decreased in the individuals with ASD compared to controls, whereas no difference was found in the mRNA expression level of MeCP2, NHE9 or AKT1. However, normalized NLGN3 and SHANK3 gene expression levels were not altered in patients with schizophrenia, and expression levels of NLGN4 and NRXN1 mRNA were not quantitatively measurable in lymphoblastoid cells. Conclusions Our results provide evidence that the NLGN3 and SHANK3 genes may be differentially expressed in lymphoblastoid cell lines from individuals with ASD compared to those from controls. These findings suggest the possibility that decreased mRNA expression levels of these genes might be involved in the pathophysiology of ASD in a substantial population of ASD patients.
Collapse
Affiliation(s)
- Yuka Yasuda
- Department of Psychiatry, Osaka University Graduate School of Medicine, D3, 2-2, Yamadaoka, Suita, 565-0871, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Proctor DT, Coulson EJ, Dodd PR. Post-synaptic scaffolding protein interactions with glutamate receptors in synaptic dysfunction and Alzheimer's disease. Prog Neurobiol 2011; 93:509-21. [PMID: 21382433 DOI: 10.1016/j.pneurobio.2011.02.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 02/18/2011] [Accepted: 02/24/2011] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease (AD) is characterized clinically by an insidious decline in cognition. Much attention has been focused on proposed pathogenic mechanisms that relate Aβ plaque and neurofibrillary tangle pathology to cognitive symptoms, but compelling evidence now identifies early synaptic loss and dysfunction, which precede plaque and tangle formation, as the more probable initiators of cognitive impairment. Glutamate-mediated transmission is severely altered in AD. Glutamate receptor expression is most markedly altered in regions of the AD brain that show the greatest pathological changes. Signaling via glutamate receptors controls synaptic strength and plasticity, and changes in these parameters are likely to contribute to memory and cognitive deficits in AD. Glutamate receptor expression and activity are modulated by interactions with post-synaptic scaffolding proteins that augment the strength and direction of signal cascades initiated by glutamate receptor activity. Scaffold proteins offer promising targets for more focused and effective drug therapy. In consequence, interest is developing into the roles these proteins play in neurological disease. In this review we discuss disruptions to excitatory neurotransmission at the level of glutamate receptor-post-synaptic scaffolding protein interactions that may contribute to synaptic dysfunction in AD.
Collapse
Affiliation(s)
- Dustin T Proctor
- School of Chemistry and Molecular Biosciences, Molecular Biosciences Building #76, Coopers Road, St Lucia campus, University of Queensland, Brisbane 4072, Australia
| | | | | |
Collapse
|
26
|
Hussman JP, Chung RH, Griswold AJ, Jaworski JM, Salyakina D, Ma D, Konidari I, Whitehead PL, Vance JM, Martin ER, Cuccaro ML, Gilbert JR, Haines JL, Pericak-Vance MA. A noise-reduction GWAS analysis implicates altered regulation of neurite outgrowth and guidance in autism. Mol Autism 2011; 2:1. [PMID: 21247446 PMCID: PMC3035032 DOI: 10.1186/2040-2392-2-1] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 01/19/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Genome-wide Association Studies (GWAS) have proved invaluable for the identification of disease susceptibility genes. However, the prioritization of candidate genes and regions for follow-up studies often proves difficult due to false-positive associations caused by statistical noise and multiple-testing. In order to address this issue, we propose the novel GWAS noise reduction (GWAS-NR) method as a way to increase the power to detect true associations in GWAS, particularly in complex diseases such as autism. METHODS GWAS-NR utilizes a linear filter to identify genomic regions demonstrating correlation among association signals in multiple datasets. We used computer simulations to assess the ability of GWAS-NR to detect association against the commonly used joint analysis and Fisher's methods. Furthermore, we applied GWAS-NR to a family-based autism GWAS of 597 families and a second existing autism GWAS of 696 families from the Autism Genetic Resource Exchange (AGRE) to arrive at a compendium of autism candidate genes. These genes were manually annotated and classified by a literature review and functional grouping in order to reveal biological pathways which might contribute to autism aetiology. RESULTS Computer simulations indicate that GWAS-NR achieves a significantly higher classification rate for true positive association signals than either the joint analysis or Fisher's methods and that it can also achieve this when there is imperfect marker overlap across datasets or when the closest disease-related polymorphism is not directly typed. In two autism datasets, GWAS-NR analysis resulted in 1535 significant linkage disequilibrium (LD) blocks overlapping 431 unique reference sequencing (RefSeq) genes. Moreover, we identified the nearest RefSeq gene to the non-gene overlapping LD blocks, producing a final candidate set of 860 genes. Functional categorization of these implicated genes indicates that a significant proportion of them cooperate in a coherent pathway that regulates the directional protrusion of axons and dendrites to their appropriate synaptic targets. CONCLUSIONS As statistical noise is likely to particularly affect studies of complex disorders, where genetic heterogeneity or interaction between genes may confound the ability to detect association, GWAS-NR offers a powerful method for prioritizing regions for follow-up studies. Applying this method to autism datasets, GWAS-NR analysis indicates that a large subset of genes involved in the outgrowth and guidance of axons and dendrites is implicated in the aetiology of autism.
Collapse
Affiliation(s)
| | - Ren-Hua Chung
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - James M Jaworski
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Daria Salyakina
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Deqiong Ma
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Ioanna Konidari
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Patrice L Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Michael L Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - John R Gilbert
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Jonathan L Haines
- Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami, 1501 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
27
|
Meldolesi J. Neurite outgrowth: this process, first discovered by Santiago Ramon y Cajal, is sustained by the exocytosis of two distinct types of vesicles. BRAIN RESEARCH REVIEWS 2011; 66:246-55. [PMID: 20600308 DOI: 10.1016/j.brainresrev.2010.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 05/31/2010] [Accepted: 06/14/2010] [Indexed: 01/26/2023]
Abstract
Neurite outgrowth is a fundamental process in the differentiation of neurons. The first, seminal study documenting the generation of "appendages" (now known as filopodia and lamellipodia) on the "cones d'accroissement," the specialized growth cones at the tips of neurites, was reported by Cajal still in the XIXth century, investigating chicken neurons embryos stained by the Golgi's reazione nera. Since then, studies have continued using, in addition to brain tissues, powerful in vitro models, i.e. primary cultures of pyramidal neurons from the hippocampus and neurosecretory cell lines, in particular PC12 cells. These studies have documented that neuronal neurites, upon sprouting from the cell body, give rise to both axons and dendrites. The specificity of these differentiated neurites depends on the diffusion barrier established at the initial segment of the axon and on the specialized domains, spines and presynaptic boutons, assembled around complexes of scaffold proteins. The two main, coordinate mechanisms that support neurite outgrowth are (a) the rearrangement of the cytoskeleton and (b) the expansion of the plasma membrane due to the exo/endocytosis of specific vesicles, distinct from those filled with neurotransmitters (clear and dense-core vesicles). The latter process is the main task of this review. In axons the surface-expanding exocytoses are concentrated at the growth cones; in dendrites they may be more distributed along the shaft. At least two types of exocytic vesicles appear to be involved, the enlargeosomes, positive for VAMP4, during early phases of development, and Ti-VAMP-positive vesicles later on. Outgrowth studies, that are now intensely pursued, have already yielded results of great importance in brain cell biology and function, and are playing an increasing role in pathology and medicine.
Collapse
Affiliation(s)
- Jacopo Meldolesi
- Department of Neuroscience, Vita-Salute San Raffaele University and San Raffaele Institute, IIT Section of Molecular Neuroscience, via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|
28
|
Previtera ML, Langhammer CG, Firestein BL. Effects of substrate stiffness and cell density on primary hippocampal cultures. J Biosci Bioeng 2010; 110:459-70. [PMID: 20547372 DOI: 10.1016/j.jbiosc.2010.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 04/01/2010] [Accepted: 04/14/2010] [Indexed: 01/06/2023]
Abstract
Previous studies have shown that dendrites are influenced by substrate stiffness when neurons are plated in either pure or mixed cultures. However, because substrate rigidity can also affect other aspects of culture development known to impact dendrite branching, such as overall cell number, it is unclear whether substrate stiffness exerts a direct or indirect effect on dendrite morphology. In this study, we determine whether substrate stiffness plays a critical role in regulating dendrite branching independent of cell number. We plated primary mixed hippocampal cultures on soft and stiff gels, with Young's moduli of 1 kPa and 7 kPa, respectively. We found that neurons plated on stiffer substrates showed increased branching relative to neurons grown on softer substrates at the same cell number. On the stiff gels, we also observed a cell number-dependent effect, in which increasing initial plating density decreased dendrite branching. This change correlates with an increase in extracellular glutamate. We concluded that both cell number and substrate stiffness play roles in determining dendrite branching, and that the two effects are independent of one another.
Collapse
Affiliation(s)
- Michelle L Previtera
- Molecular Biosciences Graduate Program, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | | | | |
Collapse
|
29
|
Microtubule stabilization by bone morphogenetic protein receptor-mediated scaffolding of c-Jun N-terminal kinase promotes dendrite formation. Mol Cell Biol 2010; 30:2241-50. [PMID: 20176805 DOI: 10.1128/mcb.01166-09] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuronal outgrowth occurs via coordinated remodeling of the cytoskeleton involving both actin and microtubules. Microtubule stabilization drives the extending neurite, yet little is known of the molecular mechanisms whereby extracellular cues regulate microtubule dynamics. Bone morphogenetic proteins (BMPs) play an important role in neuronal differentiation and morphogenesis, and BMP7 in particular induces the formation of dendrites. Here, we show that BMP7 induces stabilization of microtubules in both a MAP2-dependent neuronal cell culture model and in dendrites of primary cortical neurons. BMP7 rapidly activates c-Jun N-terminal kinases (JNKs), known regulators of microtubule dynamics, and we show that JNKs associate with the carboxy terminus of the BMP receptor, BMPRII. Activation and binding of JNKs to BMPRII is required for BMP7-induced microtubule stabilization and for BMP7-mediated dendrite formation in primary cortical neurons. These data indicate that BMPRII acts as a scaffold to localize and coordinate cytoskeletal remodeling and thereby provides an efficient means for extracellular cues, such as BMPs, to control neuronal dendritogenesis.
Collapse
|
30
|
Alié A, Manuel M. The backbone of the post-synaptic density originated in a unicellular ancestor of choanoflagellates and metazoans. BMC Evol Biol 2010; 10:34. [PMID: 20128896 PMCID: PMC2824662 DOI: 10.1186/1471-2148-10-34] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 02/03/2010] [Indexed: 11/24/2022] Open
Abstract
Background Comparative genomics of the early diverging metazoan lineages and of their unicellular sister-groups opens new window to reconstructing the genetic changes which preceded or accompanied the evolution of multicellular body plans. A recent analysis found that the genome of the nerve-less sponges encodes the homologues of most vertebrate post-synaptic proteins. In vertebrate excitatory synapses, these proteins assemble to form the post-synaptic density, a complex molecular platform linking membrane receptors, components of their signalling pathways, and the cytoskeleton. Newly available genomes from Monosiga brevicollis (a member of Choanoflagellata, the closest unicellular relatives of animals) and Trichoplax adhaerens (a member of Placozoa: besides sponges, the only nerve-less metazoans) offer an opportunity to refine our understanding of post-synaptic protein evolution. Results Searches for orthologous proteins and reconstruction of gene gains/losses based on the taxon phylogeny indicate that post-synaptic proteins originated in two main steps. The backbone scaffold proteins (Shank, Homer, DLG) and some of their partners were acquired in a unicellular ancestor of choanoflagellates and metazoans. A substantial additional set appeared in an exclusive ancestor of the Metazoa. The placozoan genome contains most post-synaptic genes but lacks some of them. Notably, the master-scaffold protein Shank might have been lost secondarily in the placozoan lineage. Conclusions The time of origination of most post-synaptic proteins was not concomitant with the acquisition of synapses or neural-like cells. The backbone of the scaffold emerged in a unicellular context and was probably not involved in cell-cell communication. Based on the reconstructed protein composition and potential interactions, its ancestral function could have been to link calcium signalling and cytoskeleton regulation. The complex later became integrated into the evolving synapse through the addition of novel functionalities.
Collapse
Affiliation(s)
- Alexandre Alié
- UPMC Univ Paris 06, UMR 7138 Systématique, Adaptation, Evolution CNRS IRD MNHN, Bâtiment A, Université Pierre et Marie Curie, 7 Quai St Bernard, 75005 Paris, France
| | | |
Collapse
|
31
|
Baucum AJ, Jalan-Sakrikar N, Jiao Y, Gustin RM, Carmody LC, Tabb DL, Ham AJL, Colbran RJ. Identification and validation of novel spinophilin-associated proteins in rodent striatum using an enhanced ex vivo shotgun proteomics approach. Mol Cell Proteomics 2010; 9:1243-59. [PMID: 20124353 DOI: 10.1074/mcp.m900387-mcp200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Spinophilin regulates excitatory postsynaptic function and morphology during development by virtue of its interactions with filamentous actin, protein phosphatase 1, and a plethora of additional signaling proteins. To provide insight into the roles of spinophilin in mature brain, we characterized the spinophilin interactome in subcellular fractions solubilized from adult rodent striatum by using a shotgun proteomics approach to identify proteins in spinophilin immune complexes. Initial analyses of samples generated using a mouse spinophilin antibody detected 23 proteins that were not present in an IgG control sample; however, 12 of these proteins were detected in complexes isolated from spinophilin knock-out tissue. A second screen using two different spinophilin antibodies and either knock-out or IgG controls identified a total of 125 proteins. The probability of each protein being specifically associated with spinophilin in each sample was calculated, and proteins were ranked according to a chi(2) analysis of the probabilities from analyses of multiple samples. Spinophilin and the known associated proteins neurabin and multiple isoforms of protein phosphatase 1 were specifically detected. Multiple, novel, spinophilin-associated proteins (myosin Va, calcium/calmodulin-dependent protein kinase II, neurofilament light polypeptide, postsynaptic density 95, alpha-actinin, and densin) were then shown to interact with GST fusion proteins containing fragments of spinophilin. Additional biochemical and transfected cell imaging studies showed that alpha-actinin and densin directly interact with residues 151-300 and 446-817, respectively, of spinophilin. Taken together, we have developed a multi-antibody, shotgun proteomics approach to characterize protein interactomes in native tissues, delineating the importance of knock-out tissue controls and providing novel insights into the nature and function of the spinophilin interactome in mature striatum.
Collapse
Affiliation(s)
- Anthony J Baucum
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232-0615, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wu SH, Arévalo JC, Sarti F, Tessarollo L, Gan WB, Chao MV. Ankyrin Repeat-rich Membrane Spanning/Kidins220 protein regulates dendritic branching and spine stability in vivo. Dev Neurobiol 2009; 69:547-57. [PMID: 19449316 PMCID: PMC4098644 DOI: 10.1002/dneu.20723] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The development of nervous system connectivity depends upon the arborization of dendritic fields and the stabilization of dendritic spine synapses. It is well established that neuronal activity and the neurotrophin BDNF modulate these correlated processes. However, the downstream mechanisms by which these extrinsic signals regulate dendritic development and spine stabilization are less well known. Here we report that a substrate of BDNF signaling, the Ankyrin Repeat-rich Membrane Spanning (ARMS) protein or Kidins220, plays a critical role in the branching of cortical and hippocampal dendrites and in the turnover of cortical spines. In the barrel somatosensory cortex and the dentate gyrus, regions where ARMS/Kidins220 is highly expressed, no difference in the complexity of dendritic arbors was observed in 1-month-old adolescent ARMS/Kidins220(+/-) mice compared to wild-type littermates. However, at 3 months of age, young adult ARMS/Kidins220(+/-) mice exhibited decreased dendritic complexity. This suggests that ARMS/Kidins220 does not play a significant role in the initial formation of dendrites but, rather, is involved in the refinement or stabilization of the arbors later in development. In addition, at 1 month of age, the rate of spine elimination was higher in ARMS/Kidins220(+/-) mice than in wild-type mice, suggesting that ARMS/Kidins220(+/-) levels regulate spine stability. Taken together, these data suggest that ARMS/Kidins220 is important for the growth of dendritic arbors and spine stability during an activity- and BDNF-dependent period of development.
Collapse
Affiliation(s)
- Synphen H. Wu
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Juan Carlos Arévalo
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Federica Sarti
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Lino Tessarollo
- Neural Development Group, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, USA
| | - Wen-Biao Gan
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Moses V. Chao
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
33
|
Chen Q, Zhu X, Zhang Y, Wetsel WC, Lee TH, Zhang X. Integrin-linked kinase is involved in cocaine sensitization by regulating PSD-95 and synapsin I expression and GluR1 Ser845 phosphorylation. J Mol Neurosci 2009; 40:284-94. [PMID: 19629758 DOI: 10.1007/s12031-009-9218-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 07/13/2009] [Indexed: 01/09/2023]
Abstract
Our recent studies have demonstrated that integrin-linked kinase (ILK) is involved in the induction and maintenance of cocaine behavioral sensitization and chronic cocaine-induced neural plasticity in the nucleus accumbens (NAc) core. In the present study, we used ILK silencing to investigate how ILK may influence cocaine-induced neural plasticity. Adeno-associated virus carrying a small interfering RNA-ILK cassette under the control of an inducible Tet-On system was injected into the NAc core of Sprague-Dawley rats. Induced silencing was established during repeated cocaine injections (sensitization induction period) or between withdrawal days 9 and 22 (sensitization maintenance period). Under both paradigms, established cocaine sensitization under non-silenced conditions was associated with enhanced PSD-95 and synapsin I protein expression as well as enhanced Ser(845) phosphorylation of the GluR1 subunit on withdrawal day. Silencing ILK expression under both paradigms prevented or reversed these changes. Importantly, ILK appears to form a complex with PSD-95 and synapsin I because it co-immunoprecipitated with each of these proteins. Together, these data suggest that ILK exerts pleiotropic actions by regulating pre- and postsynaptic neural plasticities within the NAc core in response to repeated cocaine exposure.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Box 3870, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
34
|
The Yin–Yang of Dendrite Morphology: Unity of Actin and Microtubules. Mol Neurobiol 2008; 38:270-84. [DOI: 10.1007/s12035-008-8046-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 10/07/2008] [Indexed: 10/21/2022]
|
35
|
Mukai J, Dhilla A, Drew LJ, Stark KL, Cao L, MacDermott AB, Karayiorgou M, Gogos JA. Palmitoylation-dependent neurodevelopmental deficits in a mouse model of 22q11 microdeletion. Nat Neurosci 2008; 11:1302-10. [PMID: 18836441 PMCID: PMC2756760 DOI: 10.1038/nn.2204] [Citation(s) in RCA: 209] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Accepted: 09/05/2008] [Indexed: 02/04/2023]
Abstract
Individuals with 22q11.2 microdeletions have cognitive deficits and a high risk of developing schizophrenia. Here we provide evidence that primary hippocampal neurons from a mouse model of 22q11.2 deletion (Df(16)A(+/-) mice) have decreased density of dendritic spines and glutamatergic synapses, as well as impaired dendritic growth. These deficits were prevented by introduction of the enzymatically active ZDHHC8 palmitoyltransferase encoded by a gene in the 22q11.2 locus, and they were also observed in primary cultures from Zdhhc8-deficient mice. Many of these deficits were also present in the hippocampi of adult Df(16)A(+/-) and Zdhhc8-deficient mice. Finally, we provide evidence that PSD95 is one of the substrates of ZDHHC8. Our analysis reveals that 22q11.2 microdeletion results in deficits in neuronal development and suggests that impaired neuronal protein palmitoylation contributes to many of these deficits.
Collapse
Affiliation(s)
- Jun Mukai
- Columbia University, College of Physicians & Surgeons, Department of Physiology and Cellular Biophysics, 630 West 168 Street, New York, NY 10032
| | - Alefiya Dhilla
- Columbia University, College of Physicians & Surgeons, Department of Pharmacology, 630 West 168 Street, New York, NY 10032
| | - Liam J. Drew
- Columbia University, College of Physicians & Surgeons, Department of Physiology and Cellular Biophysics, 630 West 168 Street, New York, NY 10032
| | - Kimberly L. Stark
- Columbia University, College of Physicians & Surgeons, Department of Psychiatry, 1051 Riverside Drive, New York, NY 10032
| | - Luxiang Cao
- Columbia University, College of Physicians & Surgeons, Department of Physiology and Cellular Biophysics, 630 West 168 Street, New York, NY 10032
| | - Amy B. MacDermott
- Columbia University, College of Physicians & Surgeons, Department of Physiology and Cellular Biophysics, 630 West 168 Street, New York, NY 10032
- Columbia University, College of Physicians & Surgeons, Department of Neuroscience, 1051 Riverside Drive, New York, NY 10032
| | - Maria Karayiorgou
- Columbia University, College of Physicians & Surgeons, Department of Psychiatry, 1051 Riverside Drive, New York, NY 10032
| | - Joseph A. Gogos
- Columbia University, College of Physicians & Surgeons, Department of Physiology and Cellular Biophysics, 630 West 168 Street, New York, NY 10032
- Columbia University, College of Physicians & Surgeons, Department of Neuroscience, 1051 Riverside Drive, New York, NY 10032
| |
Collapse
|