1
|
Akhgari A, Michel TM, Vafaee MS. Dendritic spines and their role in the pathogenesis of neurodevelopmental and neurological disorders. Rev Neurosci 2024; 35:489-502. [PMID: 38440811 DOI: 10.1515/revneuro-2023-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Since Cajal introduced dendritic spines in the 19th century, they have attained considerable attention, especially in neuropsychiatric and neurologic disorders. Multiple roles of dendritic spine malfunction and pathology in the progression of various diseases have been reported. Thus, it is inevitable to consider these structures as new therapeutic targets for treating neuropsychiatric and neurologic disorders such as autism spectrum disorders, schizophrenia, dementia, Down syndrome, etc. Therefore, we attempted to prepare a narrative review of the literature regarding the role of dendritic spines in the pathogenesis of aforementioned diseases and to shed new light on their pathophysiology.
Collapse
Affiliation(s)
- Aisan Akhgari
- Student Research Committee, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 5166616471, Iran
| | - Tanja Maria Michel
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Manouchehr Seyedi Vafaee
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| |
Collapse
|
2
|
Wang W, Rui M. Advances in understanding the roles of actin scaffolding and membrane trafficking in dendrite development. J Genet Genomics 2024:S1673-8527(24)00152-8. [PMID: 38925347 DOI: 10.1016/j.jgg.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Dendritic morphology is typically highly branched, and the branching and synaptic abundance of dendrites can enhance the receptive range of neurons and the diversity of information received, thus providing the basis for information processing in the nervous system. Once dendritic development is aberrantly compromised or damaged, it may lead to abnormal connectivity of the neural network, affecting the function and stability of the nervous system and ultimately triggering a series of neurological disorders. Research on the regulation of dendritic developmental processes has flourished, and much progress is now being made in its regulatory mechanisms. Noteworthily, dendrites are characterized by an extremely complex dendritic arborization that cannot be attributed to individual protein functions alone, requiring a systematic analysis of the intrinsic and extrinsic signals and the coordinated roles among them. Actin cytoskeleton organization and membrane vesicle trafficking are required during dendrite development, with actin providing tracks for vesicles and vesicle trafficking in turn providing material for actin assembly. In this review, we focus on these two basic biological processes and discuss the molecular mechanisms and their synergistic effects underlying the morphogenesis of neuronal dendrites. We also offer insights and discuss strategies for the potential preventive and therapeutic treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wanting Wang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China
| | - Menglong Rui
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China.
| |
Collapse
|
3
|
Schoonover KE, Miller NE, Fish KN, Lewis DA. Scaling of smaller pyramidal neuron size and lower energy production in schizophrenia. Neurobiol Dis 2024; 191:106394. [PMID: 38176569 PMCID: PMC10898364 DOI: 10.1016/j.nbd.2023.106394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Dorsolateral prefrontal cortex (DLPFC) dysfunction in schizophrenia appears to reflect alterations in layer 3 pyramidal neurons (L3PNs), including smaller cell bodies and lower expression of mitochondrial energy production genes. However, prior somal size studies used biased strategies for identifying L3PNs, and somal size and levels of energy production markers have not been assessed in individual L3PNs. STUDY DESIGN We combined fluorescent in situ hybridization (FISH) of vesicular glutamate transporter 1 (VGLUT1) mRNA and immunohistochemical-labeling of NeuN to determine if the cytoplasmic distribution of VGLUT1 mRNA permits the unbiased identification and somal size quantification of L3PNs. Dual-label FISH for VGLUT1 mRNA and cytochrome C oxidase subunit 4I1 (COX4I1) mRNA, a marker of energy production, was used to assess somal size and COX4I1 transcript levels in individual DLPFC L3PNs from schizophrenia (12 males; 2 females) and unaffected comparison (13 males; 1 female) subjects. STUDY RESULTS Measures of L3PN somal size with NeuN immunohistochemistry or VGLUT1 mRNA provided nearly identical results (ICC = 0.96, p < 0.0001). Mean somal size of VGLUT1-identified L3PNs was 8.7% smaller (p = 0.004) and mean COX4I1 mRNA levels per L3PN were 16.7% lower (p = 0.01) in schizophrenia. These measures were correlated across individual L3PNs in both subject groups (rrm = 0.81-0.86). CONCLUSIONS This preliminary study presents a novel method for combining unbiased neuronal identification with quantitative assessments of somal size and mRNA levels. We replicated findings of smaller somal size and lower COX4I1 mRNA levels in DLPFC L3PNs in schizophrenia. The normal scaling of COX4I1 mRNA levels with somal size in schizophrenia suggests that lower markers of energy production are secondary to L3PN morphological alterations in the illness.
Collapse
Affiliation(s)
- Kirsten E Schoonover
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Department of Psychiatry Biomedical Science Tower, W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America
| | - Nora E Miller
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Department of Psychiatry, Biomedical Science Tower W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America
| | - Kenneth N Fish
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Department of Psychiatry Biomedical Science Tower, W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Department of Psychiatry Biomedical Science Tower, W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America.
| |
Collapse
|
4
|
Malasala S, Azimian F, Chen YH, Twiss JL, Boykin C, Akhtar SN, Lu Q. Enabling Systemic Identification and Functionality Profiling for Cdc42 Homeostatic Modulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574351. [PMID: 38260445 PMCID: PMC10802479 DOI: 10.1101/2024.01.05.574351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Homeostatic modulation is pivotal in modern therapeutics. However, the discovery of bioactive materials to achieve this functionality is often random and unpredictive. Here, we enabled a systemic identification and functional classification of chemicals that elicit homeostatic modulation of signaling through Cdc42, a classical small GTPase of Ras superfamily. Rationally designed for high throughput screening, the capture of homeostatic modulators (HMs) along with molecular re-docking uncovered at least five functionally distinct classes of small molecules. This enabling led to partial agonists, hormetic agonists, bona fide activators and inhibitors, and ligand-enhanced agonists. Novel HMs exerted striking functionality in bradykinin-Cdc42 activation of actin remodelingand modified Alzheimer's disease-like behavior in mouse model. This concurrent computer-aided and experimentally empowered HM profiling highlights a model path for predicting HM landscape. One Sentence Summary With concurrent experimental biochemical profiling and in silico computer-aided drug discovery (CADD) analysis, this study enabled a systemic identification and holistic classification of Cdc42 homeostatic modulators (HMs) and demonstrated the power of CADD to predict HM classes that can mimic the pharmacological functionality of interests. Introduction Maintainingbody homeostasisis the ultimate keyto health. Thereare rich resources of bioactive materials for this functionality from both natural and synthetic chemical repertories including partial agonists (PAs) and various allosteric modulators. These homeostatic modulators (HMs) play a unique role in modern therapeutics for human diseases such as mental disorders and drug addiction. Buspirone, for example, acts as a PA for serotonin 5-HT 1A receptor but is an antagonist of the dopamine D 2 receptor. Such medical useto treat general anxietydisorders (GADs) has become one of the most-commonly prescribed medications. However, most HMs in current uses target membrane proteins and are often derived from random discoveries. HMs as therapeutics targeting cytoplasmic proteins are even more rare despite that they are in paramount needs (e. g. targeting Ras superfamily small GTPases). Rationale Cdc42, a classical member of small GTPases of Ras superfamily, regulates PI3K-AKT and Raf-MEK-ERK pathways and has been implicated in various neuropsychiatric and mental disorders as well as addictive diseases and cancer. We previously reported the high-throughput in-silico screening followed by biological characterization of novel small molecule modulators (SMMs) of Cdc42-intersectin (ITSN) protein-protein interactions (PPIs). Based on a serendipitously discovered SMM ZCL278 with PA profile as a model compound, we hypothesized that there are more varieties of such HMs of Cdc42 signaling, and the model HMs can be defined by their distinct Cdc42-ITSN binding mechanisms using computer-aided drug discovery (CADD) analysis. We further reasoned that molecular modeling coupled with experimental profiling can predict HM spectrum and thus open the door for the holistic identification and classification of multifunctional cytoplasmic target-dependent HMs as therapeutics. Results The originally discovered Cdc42 inhibitor ZCL278 displaying PA properties prompted the inquiry whether this finding represented a random encounter of PAs or whether biologically significant PAs can be widely present. The top ranked compounds were initially defined by structural fitness and binding scores to Cdc42. Because higher binding scores do not necessarily translate to higher functionality, we performed exhaustive experimentations with over 2,500 independent Cdc42-GEF (guanine nucleotide exchange factor) assays to profile the GTP loading activities on all 44 top ranked compounds derived from the SMM library. The N-MAR-GTP fluorophore-based Cdc42-GEF assay platform provided the first glimpse of the breadth of HMs. A spectrum of Cdc42 HMs was uncovered that can be categorized into five functionally distinct classes: Class I-partial competitive agonists, Class II-hormetic agonists, Class III- bona fide inhibitors (or inverse agonists), Class IV- bona fide activators or agonists, and Class V-ligand-enhanced agonists. Remarkably, model HMs such as ZCL278, ZCL279, and ZCL367 elicited striking biological functionality in bradykinin-Cdc42 activation of actin remodeling and modified Alzheimer's disease (AD)-like behavior in mouse model. Concurrently, we applied Schrödinger-enabled analyses to perform CADD predicted classification of Cdc42 HMs. We modified the classic molecular docking to instill a preferential binding pocket order (PBPO) of Cdc42-ITSN, which was based on the five binding pockets in interface of Cdc42-ITSN. We additionally applied a structure-based pharmacophore hypothesis generation for the model compounds. Then, using Schrödinger's Phase Shape, 3D ligand alignments assigned HMs to Class I, II, III, IV, and V compounds. In this HM library compounds, PBPO, matching pharmacophoric featuring, and shape alignment, all put ZCL993 in Class II compound category, which was confirmed in the Cdc42-GEF assay. Conclusion HMs can target diseased cells or tissues while minimizing impacts on tissues that are unaffected. Using Cdc42 HM model compounds as a steppingstone, GTPase activation-based screening of SMM library uncovered five functionally distinct Cdc42 HM classes among which novel efficacies towards alleviating dysregulated AD-like features in mice were identified. Furthermore, molecular re-docking of HM model compounds led to the concept of PBPO. The CADD analysis with PBPO revealed similar profile in a color-coded spectrum to these five distinct classes of Cdc42 HMs identified by biochemical functionality-based screening. The current study enabled a systemic identification and holistic classification of Cdc42 HMs and demonstrated the power of CADD to predict an HM category that can mimic the pharmacological functionality of interests. With artificial intelligence/machine learning (AI/ML) on the horizon to mirror experimental pharmacological discovery like AlphaFold for protein structure prediction, our study highlights a model path to actively capture and profile HMs in potentially any PPI landscape. Graphic Abstract Identification and functional classification of Cdc42 homeostatic modulators HMs Using Cdc42 HM model compounds as reference, GTPase activation-based screening of compound libraries uncovered five functionally distinct Cdc42 HM classes. HMs showed novel efficacies towards alleviating dysregulated Alzheimer's disease (AD)-like behavioral and molecular deficits. In parallel, molecular re-docking of HM model compounds established their preferential binding pocket orders (PBPO). PBPO-based profiling (Red reflects the most, whereas green reflects the least, preferable binding pocket) revealed trends of similar pattern to the five classes from the functionality-based classification.
Collapse
|
5
|
Tang Y, Tan Y, Palaniyappan L, Yao Y, Luo Q, Li Y. Epigenetic profile of the immune system associated with symptom severity and treatment response in schizophrenia. J Psychiatry Neurosci 2024; 49:E45-E58. [PMID: 38359932 PMCID: PMC10890792 DOI: 10.1503/jpn.230099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Environmental modification of genetic information (epigenetics) is often invoked to explain interindividual differences in the phenotype of schizophrenia. In clinical practice, such variability is most prominent in the symptom profile and the treatment response. Epigenetic regulation of immune function is of particular interest, given the therapeutic relevance of this mechanism in schizophrenia. METHODS We analyzed the DNA methylation data of immune-relevant genes in patients with schizophrenia whose disease duration was less than 3 years, with previous lifetime antipsychotic treatment of no more than 2 weeks total. RESULTS A total of 441 patients met the inclusion criteria. Core symptoms were consistently associated with 206 methylation positions, many of which had previously been implicated in inflammatory responses. Of these, 24 methylation positions were located either in regulatory regions or near the CpG islands of 20 genes, including the SRC gene, which is a key player in glutamatergic signalling. These symptom-associated immune genes were enriched in neuronal development functions, such as neuronal migration and glutamatergic synapse. Compared with using only clinical information (including scores on the Positive and Negative Syndrome Scale), integrating methylation data into the model significantly improved the predictive ability (as indicated by area under the curve) for response to 8 weeks of antipsychotic treatment. LIMITATIONS We focused on a small number of methylation probes (immune-centred search) and lacked nutritional data and direct brain-based measures. CONCLUSION Epigenetic modifications of the immune system are associated with symptom severity at onset and subsequent treatment response in schizophrenia.
Collapse
Affiliation(s)
- Yuanhao Tang
- From the National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China (Tang, Yao); the Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China (Tan, Li); the Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Que. (Palaniyappan); Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ont. (Palaniyappan); the Lawson Health Research Institute, London, Ont. (Palaniyappan); the MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (Luo)
| | - Yunlong Tan
- From the National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China (Tang, Yao); the Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China (Tan, Li); the Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Que. (Palaniyappan); Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ont. (Palaniyappan); the Lawson Health Research Institute, London, Ont. (Palaniyappan); the MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (Luo)
| | - Lena Palaniyappan
- From the National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China (Tang, Yao); the Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China (Tan, Li); the Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Que. (Palaniyappan); Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ont. (Palaniyappan); the Lawson Health Research Institute, London, Ont. (Palaniyappan); the MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (Luo)
| | - Yin Yao
- From the National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China (Tang, Yao); the Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China (Tan, Li); the Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Que. (Palaniyappan); Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ont. (Palaniyappan); the Lawson Health Research Institute, London, Ont. (Palaniyappan); the MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (Luo)
| | - Qiang Luo
- From the National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China (Tang, Yao); the Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China (Tan, Li); the Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Que. (Palaniyappan); Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ont. (Palaniyappan); the Lawson Health Research Institute, London, Ont. (Palaniyappan); the MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (Luo)
| | - Yanli Li
- From the National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China (Tang, Yao); the Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China (Tan, Li); the Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Que. (Palaniyappan); Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ont. (Palaniyappan); the Lawson Health Research Institute, London, Ont. (Palaniyappan); the MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (Luo)
| |
Collapse
|
6
|
Pordel S, Khorrami M, Saadatpour F, Rezaee D, Cho WC, Jahani S, Aghaei-Zarch SM, Hashemi E, Najafi S. The role of microRNA-185 in the pathogenesis of human diseases: A focus on cancer. Pathol Res Pract 2023; 249:154729. [PMID: 37639952 DOI: 10.1016/j.prp.2023.154729] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/29/2023] [Indexed: 08/31/2023]
Abstract
MicroRNAs (miRNAs) are a widely-studied class of non-coding RNAs characterized by their short length (18-25 nucleotides). The precise functions of miRNAs are not well-elucidated; however, an increasing number of studies suggest their involvement in various physiologic processes and deregulation in pathologic conditions. miRNA-185 (miR-185) is among the mostly-studied miRNAs in human diseases, which is found to play putative roles in conditions like metabolic disorders, asthma, frailty, schizophrenia, and hepatitis. Notably, many cancer studies report the downregulation of miR-185 in cell lines, tumor tissues, and plasma specimens of patients, while it demonstrates a suppressing role on the malignant properties of cancer cells in vitro and in vivo. Accordingly, miR-185 can be considered a tumor suppressor miRNA in human malignancies, while a few studies also report inconsistent findings. Being suggested as a prognostic/diagnostic biomarker, mi-185 is also found to offer clinical potentials, particularly for early diagnosis and prediction of the prognosis of cancer patients. In this review, we have outlined the studies that have evaluated the functions and clinical significance of miR-185 in different human diseases with a particular focus on cancer.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology and Allergy, The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Motahare Khorrami
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Saadatpour
- Pharmaceutical Biotechnology Lab, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Delsuz Rezaee
- School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| | | | - Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elham Hashemi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Tomasso MR, Padrick SB. BORG family proteins in physiology and human disease. Cytoskeleton (Hoboken) 2023; 80:182-198. [PMID: 37403807 DOI: 10.1002/cm.21768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
The binder of rho GTPases (BORG)/Cdc42 effector proteins (Cdc42EP) family is composed of five Rho GTPase binding proteins whose functions and mechanism of actions are of emerging interest. Here, we review recent findings pertaining to the family as a whole and consider how these change our understanding of cellular organization. Recent studies have implicated BORGs in both fundamental physiology and in human diseases, mainly cancers. An emerging pattern suggests that BORG family members cancer-promoting properties are related to their ability to regulate the cytoskeleton, with many impacting the organization of acto-myosin stress fibers. This is consistent with the broader literature indicating that BORG family members are regulators of both the septin and actin cytoskeleton networks. The exact mechanism through which BORGs modify the cytoskeleton is not clear, but we consider here a few data-supported and speculative possibilities. Finally, we delve into how the Rho GTPase Cdc42 modifies BORG function in cells. This remains open-ended as Cdc42's effects on BORGs appear cell type- and cell state-dependent. Collectively, these data point to the importance of the BORG family and suggest broader themes in their function and regulation.
Collapse
Affiliation(s)
- Meagan R Tomasso
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Shae B Padrick
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Mısır E, Akay GG. Synaptic dysfunction in schizophrenia. Synapse 2023:e22276. [PMID: 37210696 DOI: 10.1002/syn.22276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 04/25/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Schizophrenia is a chronic disease presented with psychotic symptoms, negative symptoms, impairment in the reward system, and widespread neurocognitive deterioration. Disruption of synaptic connections in neural circuits is responsible for the disease's development and progression. Because deterioration in synaptic connections results in the impaired effective processing of information. Although structural impairments of the synapse, such as a decrease in dendritic spine density, have been shown in previous studies, functional impairments have also been revealed with the development of genetic and molecular analysis methods. In addition to abnormalities in protein complexes regulating exocytosis in the presynaptic region and impaired vesicle release, especially, changes in proteins related to postsynaptic signaling have been reported. In particular, impairments in postsynaptic density elements, glutamate receptors, and ion channels have been shown. At the same time, effects on cellular adhesion molecular structures such as neurexin, neuroligin, and cadherin family proteins were detected. Of course, the confusing effect of antipsychotic use in schizophrenia research should also be considered. Although antipsychotics have positive and negative effects on synapses, studies indicate synaptic deterioration in schizophrenia independent of drug use. In this review, the deterioration in synapse structure and function and the effects of antipsychotics on the synapse in schizophrenia will be discussed.
Collapse
Affiliation(s)
- Emre Mısır
- Department of Psychiatry, Baskent University Faculty of Medicine, Ankara, Turkey
- Department of Interdisciplinary Neuroscience, Ankara University, Ankara, Turkey
| | - Güvem Gümüş Akay
- Department of Interdisciplinary Neuroscience, Ankara University, Ankara, Turkey
- Faculty of Medicine, Department of Physiology, Ankara University, Ankara, Turkey
- Brain Research Center (AÜBAUM), Ankara University, Ankara, Turkey
- Department of Cellular Neuroscience and Advanced Microscopic Neuroimaging, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| |
Collapse
|
9
|
Villalonga E, Mosrin C, Normand T, Girardin C, Serrano A, Žunar B, Doudeau M, Godin F, Bénédetti H, Vallée B. LIM Kinases, LIMK1 and LIMK2, Are Crucial Node Actors of the Cell Fate: Molecular to Pathological Features. Cells 2023; 12:cells12050805. [PMID: 36899941 PMCID: PMC10000741 DOI: 10.3390/cells12050805] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
LIM kinase 1 (LIMK1) and LIM kinase 2 (LIMK2) are serine/threonine and tyrosine kinases and the only two members of the LIM kinase family. They play a crucial role in the regulation of cytoskeleton dynamics by controlling actin filaments and microtubule turnover, especially through the phosphorylation of cofilin, an actin depolymerising factor. Thus, they are involved in many biological processes, such as cell cycle, cell migration, and neuronal differentiation. Consequently, they are also part of numerous pathological mechanisms, especially in cancer, where their involvement has been reported for a few years and has led to the development of a wide range of inhibitors. LIMK1 and LIMK2 are known to be part of the Rho family GTPase signal transduction pathways, but many more partners have been discovered over the decades, and both LIMKs are suspected to be part of an extended and various range of regulation pathways. In this review, we propose to consider the different molecular mechanisms involving LIM kinases and their associated signalling pathways, and to offer a better understanding of their variety of actions within the physiology and physiopathology of the cell.
Collapse
Affiliation(s)
- Elodie Villalonga
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Christine Mosrin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Thierry Normand
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Amandine Serrano
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Bojan Žunar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Michel Doudeau
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Fabienne Godin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Hélène Bénédetti
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Béatrice Vallée
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
- Correspondence: ; Tel.: +33-(0)2-38-25-76-11
| |
Collapse
|
10
|
Hinson SR, Honorat JA, Grund EM, Clarkson BD, Miske R, Scharf M, Zivelonghi C, Al-Lozi MT, Bucelli RC, Budhram A, Cho T, Choi E, Grell J, Lopez-Chiriboga AS, Levin M, Merati M, Montalvo M, Pittock SJ, Wilson MR, Howe CL, McKeon A. Septin-5 and -7-IgGs: Neurologic, Serologic, and Pathophysiologic Characteristics. Ann Neurol 2022; 92:1090-1101. [PMID: 36053822 PMCID: PMC9672904 DOI: 10.1002/ana.26482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND OBJECTIVES We sought to determine clinical significance of neuronal septin autoimmunity and evaluate for potential IgG effects. METHODS Septin-IgGs were detected by indirect immunofluorescence assays (IFAs; mouse tissue and cell based) or Western blot. IgG binding to (and internalization of) extracellular septin epitopes were evaluated for by live rat hippocampal neuron assay. The impact of purified patient IgGs on murine cortical neuron function was determined by recording extracellular field potentials in a multielectrode array platform. RESULTS Septin-IgGs were identified in 23 patients. All 8 patients with septin-5-IgG detected had cerebellar ataxia, and 7 had prominent eye movement disorders. One of 2 patients with co-existing septin-7-IgG had additional psychiatric phenotype (apathy, emotional blunting, and poor insight). Fifteen patients had septin-7 autoimmunity, without septin-5-IgG detected. Disorders included encephalopathy (11; 2 patients with accompanying myelopathy, and 2 were relapsing), myelopathy (3), and episodic ataxia (1). Psychiatric symptoms (≥1 of agitation, apathy, catatonia, disorganized thinking, and paranoia) were prominent in 6 of 11 patients with encephalopathic symptoms. Eight of 10 patients with data available (from 23 total) improved after immunotherapy, and a further 2 patients improved spontaneously. Staining of plasma membranes of live hippocampal neurons produced by patient IgGs (subclasses 1 and 2) colocalized with pre- and post-synaptic markers. Decreased spiking and bursting behavior in mixed cultures of murine glutamatergic and GABAergic cortical neurons produced by patient IgGs were attributable to neither antigenic crosslinking and internalization nor complement activation. INTERPRETATION Septin-IgGs are predictive of distinct treatment-responsive autoimmune central nervous system (CNS) disorders. Live neuron binding and induced electrophysiologic effects by patient IgGs may support septin-specific pathophysiology. ANN NEUROL 2022;92:1090-1101.
Collapse
Affiliation(s)
- Shannon R. Hinson
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | - Josephe A. Honorat
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | - Ethan M. Grund
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | | | - Ramona Miske
- Institute for Experimental Immunology, affiliated to
EUROIMMUN Medizinische Labordiagnostika, Lubeck, Germany
| | - Madeleine Scharf
- Institute for Experimental Immunology, affiliated to
EUROIMMUN Medizinische Labordiagnostika, Lubeck, Germany
| | - Cecilia Zivelonghi
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | | | | | - Adrian Budhram
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Tracey Cho
- Department of Neurology, University of Iowa, Iowa,
USA
| | - Ellie Choi
- Overlake Hospital, Bellevue, Washington, USA
| | - Jacquelyn Grell
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | | | - Marc Levin
- Department of Ophthalmology, Palo Alto Medical Foundation,
Palo Alto, CA, USA
| | - Melody Merati
- Department of Neurology, Michigan State University,
Lansing, MI, USA
| | - Mayra Montalvo
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Sean J. Pittock
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Michael R. Wilson
- Weill Institute for Neurosciences, Department of
Neurology, University of California, San Francisco, USA
| | | | - Andrew McKeon
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| |
Collapse
|
11
|
Kimoto S, Hashimoto T, Berry KJ, Tsubomoto M, Yamaguchi Y, Enwright JF, Chen K, Kawabata R, Kikuchi M, Kishimoto T, Lewis DA. Expression of actin- and oxidative phosphorylation-related transcripts across the cortical visuospatial working memory network in unaffected comparison and schizophrenia subjects. Neuropsychopharmacology 2022; 47:2061-2070. [PMID: 35034100 PMCID: PMC9556568 DOI: 10.1038/s41386-022-01274-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 11/09/2022]
Abstract
Visuospatial working memory (vsWM), which is impaired in schizophrenia (SZ), is mediated by a distributed cortical network. In one node of this network, the dorsolateral prefrontal cortex (DLPFC), altered expression of transcripts for actin assembly and mitochondrial oxidative phosphorylation (OXPHOS) have been reported in SZ. To understand the relationship between these processes, and the extent to which similar alterations are present in other regions of vsWM network in SZ, a subset of actin- (CDC42, BAIAP2, ARPC3, and ARPC4) and OXPHOS-related (ATP5H, COX4I1, COX7B, and NDUFB3) transcripts were quantified in DLPFC by RNA sequencing in 139 SZ and unaffected comparison (UC) subjects, and in DLPFC and three other regions of the cortical vsWM network by qPCR in 20 pairs of SZ and UC subjects. By RNA sequencing, levels of actin- and OXPHOS-related transcripts were significantly altered in SZ, and robustly correlated in both UC and SZ subject groups. By qPCR, cross-regional expression patterns of these transcripts in UC subjects were consistent with greater actin assembly in DLPFC and higher OXPHOS activity in primary visual cortex (V1). In SZ, CDC42 and ARPC4 levels were lower in all regions, BAIAP2 levels higher only in V1, and ARPC3 levels unaltered across regions. All OXPHOS-related transcript levels were lower in SZ, with the disease effect decreasing from posterior to anterior regions. The differential alterations in markers of actin assembly and energy production across regions of the cortical vsWM network in SZ suggest that each region may make specific contributions to vsWM impairments in the illness.
Collapse
Affiliation(s)
- Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - Takanori Hashimoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Research Center for Child Development, Kanazawa University, Kanazawa, 920-8640, Japan
| | - Kimberly J Berry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Makoto Tsubomoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yasunari Yamaguchi
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - John F Enwright
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kehui Chen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Research Center for Child Development, Kanazawa University, Kanazawa, 920-8640, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
12
|
Dienel SJ, Schoonover KE, Lewis DA. Cognitive Dysfunction and Prefrontal Cortical Circuit Alterations in Schizophrenia: Developmental Trajectories. Biol Psychiatry 2022; 92:450-459. [PMID: 35568522 PMCID: PMC9420748 DOI: 10.1016/j.biopsych.2022.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 01/01/2023]
Abstract
Individuals with schizophrenia (SZ) exhibit cognitive performance below expected levels based on familial cognitive aptitude. One such cognitive process, working memory (WM), is robustly impaired in SZ. These WM impairments, which emerge over development during the premorbid and prodromal stages of SZ, appear to reflect alterations in the neural circuitry of the dorsolateral prefrontal cortex. Within the dorsolateral prefrontal cortex, a microcircuit formed by reciprocal connections between excitatory layer 3 pyramidal neurons and inhibitory parvalbumin basket cells (PVBCs) appears to be a key neural substrate for WM. Postmortem human studies indicate that both layer 3 pyramidal neurons and PVBCs are altered in SZ, suggesting that levels of excitation and inhibition are lower in the microcircuit. Studies in monkeys indicate that features of both cell types exhibit distinctive postnatal developmental trajectories. Together, the results of these studies suggest a model in which 1) genetic and/or early environmental insults to excitatory signaling in layer 3 pyramidal neurons give rise to cognitive impairments during the prodromal phase of SZ and evoke compensatory changes in inhibition that alter the developmental trajectories of PVBCs, and 2) synaptic pruning during adolescence further lowers excitatory activity to a level that exceeds the compensatory capacity of PVBC inhibition, leading to a failure of the normal maturational improvements in WM during the prodromal and early clinical stages of SZ. Findings that support as well as challenge this model are discussed.
Collapse
Affiliation(s)
- Samuel J Dienel
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kirsten E Schoonover
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
13
|
Byeon S, Werner B, Falter R, Davidsen K, Snyder C, Ong SE, Yadav S. Proteomic Identification of Phosphorylation-Dependent Septin 7 Interactors that Drive Dendritic Spine Formation. Front Cell Dev Biol 2022; 10:836746. [PMID: 35602601 PMCID: PMC9114808 DOI: 10.3389/fcell.2022.836746] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Septins are a family of cytoskeletal proteins that regulate several important aspects of neuronal development. Septin 7 (Sept7) is enriched at the base of dendritic spines in excitatory neurons and mediates both spine formation and spine and synapse maturation. Phosphorylation at a conserved C-terminal tail residue of Sept7 mediates its translocation into the dendritic spine head to allow spine and synapse maturation. The mechanistic basis for postsynaptic stability and compartmentalization conferred by phosphorylated Sept7, however, is unclear. We report herein the proteomic identification of Sept7 phosphorylation-dependent neuronal interactors. Using Sept7 C-terminal phosphopeptide pulldown and biochemical assays, we show that the 14-3-3 family of proteins specifically interacts with Sept7 when phosphorylated at the T426 residue. Biochemically, we validate the interaction between Sept7 and 14-3-3 isoform gamma and show that 14-3-3 gamma is also enriched in the mature dendritic spine head. Furthermore, we demonstrate that interaction of phosphorylated Sept7 with 14-3-3 protects it from dephosphorylation, as expression of a 14-3-3 antagonist significantly decreases phosphorylated Sept7 in neurons. This study identifies 14-3-3 proteins as an important physiological regulator of Sept7 function in neuronal development.
Collapse
Affiliation(s)
- Sujin Byeon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Reilly Falter
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Kristian Davidsen
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Calvin Snyder
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
14
|
RhoA Signaling in Neurodegenerative Diseases. Cells 2022; 11:cells11091520. [PMID: 35563826 PMCID: PMC9103838 DOI: 10.3390/cells11091520] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
Ras homolog gene family member A (RhoA) is a small GTPase of the Rho family involved in regulating multiple signal transduction pathways that influence a diverse range of cellular functions. RhoA and many of its downstream effector proteins are highly expressed in the nervous system, implying an important role for RhoA signaling in neurons and glial cells. Indeed, emerging evidence points toward a role of aberrant RhoA signaling in neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. In this review, we summarize the current knowledge of RhoA regulation and downstream cellular functions with an emphasis on the role of RhoA signaling in neurodegenerative diseases and the therapeutic potential of RhoA inhibition in neurodegeneration.
Collapse
|
15
|
Ying L, Zhao J, Ye Y, Liu Y, Xiao B, Xue T, Zhu H, Wu Y, He J, Qin S, Jiang Y, Guo F, Zhang L, Liu N, Zhang L. Regulation of Cdc42 signaling by the dopamine D2 receptor in a mouse model of Parkinson's disease. Aging Cell 2022; 21:e13588. [PMID: 35415964 PMCID: PMC9124300 DOI: 10.1111/acel.13588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/29/2022] [Accepted: 02/26/2022] [Indexed: 12/02/2022] Open
Abstract
Substantial spine loss in striatal medium spiny neurons (MSNs) and abnormal behaviors are common features of Parkinson's disease (PD). The caudate putamen (CPu) mainly contains MSNs expressing dopamine D1 receptor (dMSNs) and dopamine D2 receptor (iMSNs) exerting critical effects on motor and cognition behavior. However, the molecular mechanisms contributing to spine loss and abnormal behaviors in dMSNs and iMSNs under parkinsonian state remain unknown. In the present study, we revealed that Cell division control protein 42 (Cdc42) signaling was significantly decreased in the caudate putamen (CPu) in parkinsonian mice. In addition, overexpression of constitutively active Cdc42 in the CPu reversed spine abnormalities and improved the behavior deficits in parkinsonian mice. Utilizing conditional dopamine D1 receptor (D1R) or D2 receptor (D2R) knockout mice, we found that such a decrease under parkinsonian state was further reduced by conditional knockout of the D2R but not D1R. Moreover, the thin spine loss in iMSNs and deficits in motor coordination and cognition induced by conditional knockout of D2R were reversed by overexpression of constitutively active Cdc42 in the CPu. Additionally, conditional knockout of Cdc42 from D2R‐positive neurons in the CPu was sufficient to induce spine and behavior deficits similar to those observed in parkinsonian mice. Overall, our results indicate that impaired Cdc42 signaling regulated by D2R plays an important role in spine loss and behavioral deficits in PD.
Collapse
Affiliation(s)
- Li Ying
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Jinlan Zhao
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yingshan Ye
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yutong Liu
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Bin Xiao
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Tao Xue
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Hangfei Zhu
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yue Wu
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Jing He
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Sifei Qin
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yong Jiang
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology Children's Hospital Research Foundation Cincinnati Ohio USA
| | - Lin Zhang
- Department of Histology and Embryology NMPA Key Laboratory for Safety Evaluation of Cosmetics Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province School of Basic Medical Sciences Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Nuyun Liu
- Laboratory Animal Center Elderly Health Services Research Center Southern Medical University Guangzhou China
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| |
Collapse
|
16
|
Jiao R, Chen X, Boerwinkle E, Xiong M. Genome-Wide Causation Studies of Complex Diseases. J Comput Biol 2022; 29:908-931. [PMID: 35451855 DOI: 10.1089/cmb.2021.0676] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite significant progress in dissecting the genetic architecture of complex diseases by genome-wide association studies (GWAS), the signals identified by association analysis may not have specific pathological relevance to diseases so that a large fraction of disease-causing genetic variants is still hidden. Association is used to measure dependence between two variables or two sets of variables. GWAS test association between a disease and single-nucleotide polymorphisms (SNPs) (or other genetic variants) across the genome. Association analysis may detect superficial patterns between disease and genetic variants. Association signals provide limited information on the causal mechanism of diseases. The use of association analysis as a major analytical platform for genetic studies of complex diseases is a key issue that may hamper discovery of disease mechanisms, calling into the questions the ability of GWAS to identify loci-underlying diseases. It is time to move beyond association analysis toward techniques, which enables the discovery of the underlying causal genetic structures of complex diseases. To achieve this, we propose the concept of genome-wide causation studies (GWCS) as an alternative to GWAS and develop additive noise models (ANMs) for genetic causation analysis. Type 1 error rates and power of the ANMs in testing causation are presented. We conducted GWCS of schizophrenia. Both simulation and real data analysis show that the proportion of the overlapped association and causation signals is small. Thus, we anticipate that our analysis will stimulate serious discussion of the applicability of GWAS and GWCS.
Collapse
Affiliation(s)
- Rong Jiao
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiangning Chen
- Department of Psychology, Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Nevada, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Momiao Xiong
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
17
|
Sabaie H, Gharesouran J, Asadi MR, Farhang S, Ahangar NK, Brand S, Arsang-Jang S, Dastar S, Taheri M, Rezazadeh M. Downregulation of miR-185 is a common pathogenic event in 22q11.2 deletion syndrome-related and idiopathic schizophrenia. Metab Brain Dis 2022; 37:1175-1184. [PMID: 35075501 DOI: 10.1007/s11011-022-00918-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
Abstract
Schizophrenia (SCZ) is known as a complicated mental disease with an unknown etiology. The microdeletion of 22q11.2 is the most potent genetic risk factor. Researchers are still trying to find which genes in the deletion region are linked to SCZ. MIR185, encoding microRNA (miR)-185, is present in the minimal 1.5 megabase deletion. Nonetheless, the miR-185 expression profile and its corresponding target genes in animal models and patients with 22q11.2 deletion syndrome (22q11.2DS) imply that more study is required about miR-185 and its corresponding downstream pathways within idiopathic SCZ. The expression of hsa-miR-185-5p and its corresponding target gene, shisa family member 7 (SHISA7), sometimes called CKAMP59, were evaluated in the peripheral blood (PB) samples of Iranian Azeri patients with idiopathic SCZ and healthy subjects, matched by gender and age as control groups by quantitative polymerase chain reaction (qPCR). Fifty SCZ patients (male/female: 22/28, age (mean ± standard deviation (SD)): 35.9 ± 5.6) and 50 matched healthy controls (male/female: 23/27, age (mean ± SD): 34.7 ± 5.4) were enrolled. The expression of hsa-miR-185-5p in the PB samples from subjects with idiopathic SCZ was substantially lower than in that of control groups (posterior beta = -0.985, adjusted P-value < 0.0001). There was also a difference within the expression profile between female and male subgroups (posterior beta = -0.86, adjusted P-value = 0.046 and posterior beta = -1.015, adjusted P-value = 0.004, in turn). Nevertheless, no significant difference was present in the expression level of CKAMP59 between PB samples from patients and control groups (adjusted P-value > 0.999). The analysis of the receiver operating characteristic (ROC) curve suggested that hsa-miR-185-5p may correctly distinguish subjects with idiopathic SCZ from healthy people (the area under curve (AUC) value: 0.722). Furthermore, there was a strong positive correlation between the expression pattern of the abovementioned genes in patients with SCZ and healthy subjects (r = 0.870, P < 0.001 and r = 0.812, P < 0.001, respectively), indicating that this miR works as an enhancer. More research is needed to determine if the hsa-miR-185-5p has an enhancer activity. In summary, this is the first research to highlight the expression of the miR-185 and CKAMP59 genes in the PB from subjects with idiopathic SCZ. Our findings suggest that gene expression alterations mediated by miR-185 may play a role in the pathogenesis of idiopathic and 22q11.2DS SCZ. It is worth noting that, despite a substantial and clear relationship between CKAMP59 and hsa-miR-185-5p, indicating an interactive network, their involvement in the development of SCZ should be reconsidered based on the whole blood sample since the changed expression level of CKAMP59 was not significant. Further research with greater sample sizes and particular leukocyte subsets can greatly make these results stronger.
Collapse
Affiliation(s)
- Hani Sabaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Gharesouran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Asadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Farhang
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Rob Giel Research Center, University Medical Center Groningen, University Center for Psychiatry, University of Groningen, Groningen, Netherlands
| | - Noora Karim Ahangar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Serge Brand
- Psychiatric Clinics, Center for Affective, Stress and Sleep Disorders, University of Basel, Basel, Switzerland
| | - Shahram Arsang-Jang
- Cancer Gene Therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran
| | - Saba Dastar
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Fatih, Istanbul, Turkey
| | - Mohammad Taheri
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Rezazadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Disruption of Alternative Splicing in the Amygdala of Pigs Exposed to Maternal Immune Activation. IMMUNO 2021. [DOI: 10.3390/immuno1040035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response of gestating females to infection or stress can disrupt gene expression in the offspring’s amygdala, resulting in lasting neurodevelopmental, physiological, and behavioral disorders. The effects of maternal immune activation (MIA) can be impacted by the offspring’s sex and exposure to additional stressors later in life. The objectives of this study were to investigate the disruption of alternative splicing patterns associated with MIA in the offspring’s amygdala and characterize this disruption in the context of the second stress of weaning and sex. Differential alternative splicing was tested on the RNA-seq profiles of a pig model of viral-induced MIA. Compared to controls, MIA was associated with the differential alternative splicing (FDR-adjusted p-value < 0.1) of 292 and 240 genes in weaned females and males, respectively, whereas 132 and 176 genes were differentially spliced in control nursed female and male, respectively. The majority of the differentially spliced (FDR-adjusted p-value < 0.001) genes (e.g., SHANK1, ZNF672, KCNA6) and many associated enriched pathways (e.g., Fc gamma R-mediated phagocytosis, non-alcoholic fatty liver disease, and cGMP-PKG signaling) have been reported in MIA-related disorders including autism and schizophrenia in humans. Differential alternative splicing associated with MIA was detected in the gene MAG across all sex-stress groups except for unstressed males and SLC2A11 across all groups except unstressed females. Precise understanding of the effect of MIA across second stressors and sexes necessitates the consideration of splicing isoform profiles.
Collapse
|
19
|
Shen L, Lv X, Huang H, Li M, Huai C, Wu X, Wu H, Ma J, Chen L, Wang T, Tan J, Sun Y, Li L, Shi Y, Yang C, Cai L, Lu Y, Zhang Y, Weng S, Tai S, Zhang N, He L, Wan C, Qin S. Genome-wide analysis of DNA methylation in 106 schizophrenia family trios in Han Chinese. EBioMedicine 2021; 72:103609. [PMID: 34628353 PMCID: PMC8511801 DOI: 10.1016/j.ebiom.2021.103609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/05/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022] Open
Abstract
Background Schizophrenia (SCZ) is a severe psychiatric disorder that affects approximately 0.75% of the global population. Both genetic and environmental factors contribute to development of SCZ. SCZ tends to run in family while both genetic and environmental factor contribute to its etiology. Much evidence suggested that alterations in DNA methylations occurred in SCZ patients. Methods To investigate potential inheritable pattern of DNA methylation in SCZ family, we performed a genome-wide analysis of DNA methylation of peripheral blood samples from 106 Chinese SCZ family trios. Genome-wide DNA methylations were quantified by Agilent 1 × 244 k Human Methylation Microarray. Findings In this study, we proposed a loci inheritance frequency model that allows characterization of differential methylated regions as SCZ biomarkers. Based on this model, 112 hypermethylated and 125 hypomethylated regions were identified. Additionally, 121 hypermethylated and 139 hypomethylated genes were annotated. The results of functional enrichment analysis indicated that multiple differentially methylated genes (DMGs) involved in Notch/HH/Wnt signaling, MAPK signaling, GPCR signaling, immune response signaling. Notably, a number of hypomethylated genes were significantly enriched in cerebral cortex and functionally enriched in nervous system development. Interpretation Our findings not only validated previously discovered risk genes of SCZ but also identified novel candidate DMGs in SCZ. These results may further the understanding of altered DNA methylations in SCZ.
Collapse
Affiliation(s)
- Lu Shen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoying Lv
- DCH Technologies Inc, Cambridge, MA 02142, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, Shanghai 200031, PR China; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mo Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Cong Huai
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Xi Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Hao Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Jingsong Ma
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Luan Chen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Ting Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Jie Tan
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yidan Sun
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lixing Li
- Department of General Surgery, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Shi
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Chao Yang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lei Cai
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yana Lu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi 214151, China
| | - Yan Zhang
- The Second People's Hospital of Lishui, Lishui 323020, China
| | - Saizheng Weng
- Fuzhou Neuro-psychiatric hospital, Fujian Medical University, Fuzhou 350026, China
| | - Shaobin Tai
- The Second People's Hospital of Huangshan, Huangshan 245021, China
| | - Na Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lin He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China; Department of General Surgery, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Chunling Wan
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| | - Shengying Qin
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| |
Collapse
|
20
|
Namme JN, Bepari AK, Takebayashi H. Cofilin Signaling in the CNS Physiology and Neurodegeneration. Int J Mol Sci 2021; 22:ijms221910727. [PMID: 34639067 PMCID: PMC8509315 DOI: 10.3390/ijms221910727] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022] Open
Abstract
All eukaryotic cells are composed of the cytoskeleton, which plays crucial roles in coordinating diverse cellular functions such as cell division, morphology, migration, macromolecular stabilization, and protein trafficking. The cytoskeleton consists of microtubules, intermediate filaments, and actin filaments. Cofilin, an actin-depolymerizing protein, is indispensable for regulating actin dynamics in the central nervous system (CNS) development and function. Cofilin activities are spatiotemporally orchestrated by numerous extra- and intra-cellular factors. Phosphorylation at Ser-3 by kinases attenuate cofilin’s actin-binding activity. In contrast, dephosphorylation at Ser-3 enhances cofilin-induced actin depolymerization. Cofilin functions are also modulated by various binding partners or reactive oxygen species. Although the mechanism of cofilin-mediated actin dynamics has been known for decades, recent research works are unveiling the profound impacts of cofilin dysregulation in neurodegenerative pathophysiology. For instance, oxidative stress-induced increase in cofilin dephosphorylation is linked to the accumulation of tau tangles and amyloid-beta plaques in Alzheimer’s disease. In Parkinson’s disease, cofilin activation by silencing its upstream kinases increases α-synuclein-fibril entry into the cell. This review describes the molecular mechanism of cofilin-mediated actin dynamics and provides an overview of cofilin’s importance in CNS physiology and pathophysiology.
Collapse
Affiliation(s)
- Jannatun Nayem Namme
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
| | - Asim Kumar Bepari
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
- Correspondence: (A.K.B.); (H.T.)
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- Correspondence: (A.K.B.); (H.T.)
| |
Collapse
|
21
|
Woo E, Sansing LH, Arnsten AFT, Datta D. Chronic Stress Weakens Connectivity in the Prefrontal Cortex: Architectural and Molecular Changes. CHRONIC STRESS 2021; 5:24705470211029254. [PMID: 34485797 PMCID: PMC8408896 DOI: 10.1177/24705470211029254] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022]
Abstract
Chronic exposure to uncontrollable stress causes loss of spines and dendrites in the prefrontal cortex (PFC), a recently evolved brain region that provides top-down regulation of thought, action, and emotion. PFC neurons generate top-down goals through recurrent excitatory connections on spines. This persistent firing is the foundation for higher cognition, including working memory, and abstract thought. However, exposure to acute uncontrollable stress drives high levels of catecholamine release in the PFC, which activates feedforward calcium-cAMP signaling pathways to open nearby potassium channels, rapidly weakening synaptic connectivity to reduce persistent firing. Chronic stress exposures can further exacerbate these signaling events leading to loss of spines and resulting in marked cognitive impairment. In this review, we discuss how stress signaling mechanisms can lead to spine loss, including changes to BDNF-mTORC1 signaling, calcium homeostasis, actin dynamics, and mitochondrial actions that engage glial removal of spines through inflammatory signaling. Stress signaling events may be amplified in PFC spines due to cAMP magnification of internal calcium release. As PFC dendritic spine loss is a feature of many cognitive disorders, understanding how stress affects the structure and function of the PFC will help to inform strategies for treatment and prevention.
Collapse
Affiliation(s)
- Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA.,Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Lauren H Sansing
- Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| |
Collapse
|
22
|
Chen W, Zhu Y, Zhang W, Zhang H, Zhou Y, Sun P, Wu G. CDC42EP3 is a key promoter involved in the development and progression of gastric cancer. Carcinogenesis 2021; 42:1179-1188. [PMID: 34111280 DOI: 10.1093/carcin/bgab048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/19/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent cancers and severely endangers human health. Due to the low rate of diagnosis, most patients with gastric cancer are diagnosed as advanced. CDC42 effector protein 3 (CDC42EP3) has been revealed to be involved in several types of human cancers' development and progression. However, the function of CDC42EP3 in GC is not yet clear. CDC42EP3 expression was detected by immunohistochemistry (IHC), qRT-PCR and Western blot assay in tumor tissues and cell lines of GC. CDC42EP3 knockdown cell models were constructed by lentivirus transfection. Cell proliferation was evaluated by the MTT assay. The wound-healing assay and the transwell assay were utilized to assess the cell migration. Also, the cell apoptosis and the cell cycle were evaluated by flow cytometry. Moreover, the mechanism was investigated by Human Apoptosis Antibody Array. The in vivo experiments were conducted to verify the effects of CDC42EP3 knockdown on the tumor growth of GC. The expression level of CDC42EP3 was up-regulated in tumor tissues. High CDC42EP3 expression was positively related to more advanced tumor grade. CDC42EP3 knockdown inhibited cell proliferation and migration, promoted cell apoptosis and suppressed the tumor growth. On the other hand, it was also found that the silencing of CDC42EP3 inhibited HSP27 and IGF-1sR expression as well as promoted Caspase3, p53, TNF-α, TNF-β, TRAILR-1 and TRAILR-2 expression. CDC42EP3 was revealed to work as a tumor promoter in the development and progression of GC, which could be a promising therapeutic target for the therapy of GC.
Collapse
Affiliation(s)
- Wenchao Chen
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yuanzeng Zhu
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Wei Zhang
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Han Zhang
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yang Zhou
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Peichun Sun
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Gang Wu
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
23
|
Parellada E, Gassó P. Glutamate and microglia activation as a driver of dendritic apoptosis: a core pathophysiological mechanism to understand schizophrenia. Transl Psychiatry 2021; 11:271. [PMID: 33958577 PMCID: PMC8102516 DOI: 10.1038/s41398-021-01385-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Schizophrenia disorder remains an unsolved puzzle. However, the integration of recent findings from genetics, molecular biology, neuroimaging, animal models and translational clinical research offers evidence that the synaptic overpruning hypothesis of schizophrenia needs to be reassessed. During a critical period of neurodevelopment and owing to an imbalance of excitatory glutamatergic pyramidal neurons and inhibitory GABAergic interneurons, a regionally-located glutamate storm might occur, triggering excessive dendritic pruning with the activation of local dendritic apoptosis machinery. The apoptotic loss of dendritic spines would be aggravated by microglia activation through a recently described signaling system from complement abnormalities and proteins of the MHC, thus implicating the immune system in schizophrenia. Overpruning of dendritic spines coupled with aberrant synaptic plasticity, an essential function for learning and memory, would lead to brain misconnections and synaptic inefficiency underlying the primary negative symptoms and cognitive deficits of schizophrenia. This driving hypothesis has relevant therapeutic implications, including the importance of pharmacological interventions during the prodromal phase or the transition to psychosis, targeting apoptosis, microglia cells or the glutamate storm. Future research on apoptosis and brain integrity should combine brain imaging, CSF biomarkers, animal models and cell biology.
Collapse
Affiliation(s)
- Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU). Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Catalonia, Spain.
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| | - Patricia Gassó
- Barcelona Clínic Schizophrenia Unit (BCSU). Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
24
|
Martínez-Torres NI, Vázquez-Hernández N, Martín-Amaya-Barajas FL, Flores-Soto M, González-Burgos I. Ibotenic acid induced lesions impair the modulation of dendritic spine plasticity in the prefrontal cortex and amygdala, a phenomenon that underlies working memory and social behavior. Eur J Pharmacol 2021; 896:173883. [PMID: 33513334 DOI: 10.1016/j.ejphar.2021.173883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/08/2023]
Abstract
The lesions induced by Ibotenic acid (IA) emulate some of the symptoms associated with schizophrenia, such as impaired working memory that is predominantly organized by the medial prefrontal cortex (mPFC), or difficulties in social interactions that aremainly organized by the amygdala (AMG). The plastic capacity of dendritic spines in neurons of the mPFC and AMG is modulated by molecules that participate in the known deterioration of working memory, although the influence of these on the socialization of schizophrenic patients is unknown. Here, the effect of a neonatal IA induced lesion on social behavior and working memory was evaluated in adult rats, along with the changes in cytoarchitecture of dendritic spines and their protein content, specifically the postsynaptic density protein 95 (PSD-95), Synaptophysin (Syn), AMPA receptors, and brain-derived neurotrophic factor (BDNF). Both working memory and social behavior were impaired, and the density of the spines, as well as their PSD-95, Syn, AMPA receptor and BDNF content was lower in IA lesioned animals. The proportional density of thin, mushroom, stubby and wide spines resulted in plastic changes that suggest the activation of compensatory processes in the face of the adverse effects of the lesion. In addition, the reduction in the levels of the modulating factors also suggests that the signaling pathways in which such factors are implicated would be altered in the brains of patients with schizophrenia. Accordingly, the experimental study of such signaling pathways is likely to aid the development of more effective pharmacological strategies for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Néstor I Martínez-Torres
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico; Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jal., Mexico
| | - Nallely Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico
| | | | - Mario Flores-Soto
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico
| | - Ignacio González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico.
| |
Collapse
|
25
|
Qin X, Chen J, Zhou T. 22q11.2 deletion syndrome and schizophrenia. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1181-1190. [PMID: 33098288 DOI: 10.1093/abbs/gmaa113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
22q11.2 deletion is a common microdeletion that causes an array of developmental defects including 22q11.2 deletion syndrome (22q11DS) or DiGeorge syndrome and velocardiofacial syndrome. About 30% of patients with 22q11.2 deletion develop schizophrenia. Mice with deletion of the ortholog region in mouse chromosome 16qA13 exhibit schizophrenia-like abnormal behaviors. It is suggested that the genes deleted in 22q11DS are involved in the pathogenesis of schizophrenia. Among these genes, COMT, ZDHHC8, DGCR8, and PRODH have been identified as schizophrenia susceptibility genes. And DGCR2 is also found to be associated with schizophrenia. In this review, we focused on these five genes and reviewed their functions in the brain and the potential pathophysiological mechanisms in schizophrenia, which will give us a deeper understanding of the pathology of schizophrenia.
Collapse
Affiliation(s)
- Xianzheng Qin
- Queen Mary School of Nanchang University, Nanchang University, Nanchang 330031, China
| | - Jiang Chen
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| |
Collapse
|
26
|
Zhang Z, Ye M, Li Q, You Y, Yu H, Ma Y, Mei L, Sun X, Wang L, Yue W, Li R, Li J, Zhang D. The Schizophrenia Susceptibility Gene OPCML Regulates Spine Maturation and Cognitive Behaviors through Eph-Cofilin Signaling. Cell Rep 2020; 29:49-61.e7. [PMID: 31577955 DOI: 10.1016/j.celrep.2019.08.091] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 07/09/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022] Open
Abstract
Previous genetic and biological evidence converge on the involvement of synaptic dysfunction in schizophrenia, and OPCML, encoding a synaptic membrane protein, is reported to be genetically associated with schizophrenia. However, its role in the pathophysiology of schizophrenia remains largely unknown. Here, we found that Opcml is strongly expressed in the mouse hippocampus; ablation of Opcml leads to reduced phosphorylated cofilin and dysregulated F-actin dynamics, which disturbs the spine maturation. Furthermore, Opcml interacts with EphB2 to control the stability of spines by regulating the ephrin-EphB2-cofilin signaling pathway. Opcml-deficient mice display impaired cognitive behaviors and abnormal sensorimotor gating, which are similar to features in neuropsychiatric disorders such as schizophrenia. Notably, the administration of aripiprazole partially restores the abnormal behaviors in Opcml-/- mice by increasing the phosphorylated cofilin level and facilitating spine maturation. We demonstrated a critical role of the schizophrenia-susceptible gene OPCML in spine maturation and cognitive behaviors via regulating the ephrin-EphB2-cofilin signaling pathway, providing further insights into the characteristics of schizophrenia.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Maoqing Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Qiongwei Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yang You
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Hao Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yuanlin Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Liwei Mei
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xiaqin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Lifang Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Rena Li
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jun Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Dai Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
27
|
Schill Y, Bijata M, Kopach O, Cherkas V, Abdel-Galil D, Böhm K, Schwab MH, Matsuda M, Compan V, Basu S, Bijata K, Wlodarczyk J, Bard L, Cole N, Dityatev A, Zeug A, Rusakov DA, Ponimaskin E. Serotonin 5-HT 4 receptor boosts functional maturation of dendritic spines via RhoA-dependent control of F-actin. Commun Biol 2020; 3:76. [PMID: 32060357 PMCID: PMC7021812 DOI: 10.1038/s42003-020-0791-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/23/2020] [Indexed: 01/24/2023] Open
Abstract
Activity-dependent remodeling of excitatory connections underpins memory formation in the brain. Serotonin receptors are known to contribute to such remodeling, yet the underlying molecular machinery remains poorly understood. Here, we employ high-resolution time-lapse FRET imaging in neuroblastoma cells and neuronal dendrites to establish that activation of serotonin receptor 5-HT4 (5-HT4R) rapidly triggers spatially-restricted RhoA activity and G13-mediated phosphorylation of cofilin, thus locally boosting the filamentous actin fraction. In neuroblastoma cells, this leads to cell rounding and neurite retraction. In hippocampal neurons in situ, 5-HT4R-mediated RhoA activation triggers maturation of dendritic spines. This is paralleled by RhoA-dependent, transient alterations in cell excitability, as reflected by increased spontaneous synaptic activity, apparent shunting of evoked synaptic responses, and enhanced long-term potentiation of excitatory transmission. The 5-HT4R/G13/RhoA signaling thus emerges as a previously unrecognized molecular pathway underpinning use-dependent functional remodeling of excitatory synaptic connections.
Collapse
Affiliation(s)
- Yvonne Schill
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Monika Bijata
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Olga Kopach
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Volodymyr Cherkas
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Dalia Abdel-Galil
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Katrin Böhm
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Markus H Schwab
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Michiyuki Matsuda
- Bioimaging and Cell Signaling, Kyoto University, Kyoto, 606-8501, Japan
| | | | - Subhadip Basu
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
- Computer Science and Engineering, Jadavpur University, Kolkata, 700032, India
| | - Krystian Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Jakub Wlodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Lucie Bard
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Nicholas Cole
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Dmitri A Rusakov
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
28
|
Pinner AL, Mueller TM, Alganem K, McCullumsmith R, Meador-Woodruff JH. Protein expression of prenyltransferase subunits in postmortem schizophrenia dorsolateral prefrontal cortex. Transl Psychiatry 2020; 10:3. [PMID: 32066669 PMCID: PMC7026430 DOI: 10.1038/s41398-019-0610-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 09/09/2019] [Accepted: 10/03/2019] [Indexed: 12/22/2022] Open
Abstract
The pathophysiology of schizophrenia includes altered neurotransmission, dysregulated intracellular signaling pathway activity, and abnormal dendritic morphology that contribute to deficits of synaptic plasticity in the disorder. These processes all require dynamic protein-protein interactions at cell membranes. Lipid modifications target proteins to membranes by increasing substrate hydrophobicity by the addition of a fatty acid or isoprenyl moiety, and recent evidence suggests that dysregulated posttranslational lipid modifications may play a role in multiple neuropsychiatric disorders, including schizophrenia. Consistent with these emerging findings, we have recently reported decreased protein S-palmitoylation in schizophrenia. Protein prenylation is a lipid modification that occurs upstream of S-palmitoylation on many protein substrates, facilitating membrane localization and activity of key intracellular signaling proteins. Accordingly, we hypothesized that, in addition to palmitoylation, protein prenylation may be abnormal in schizophrenia. To test this, we assayed protein expression of the five prenyltransferase subunits (FNTA, FNTB, PGGT1B, RABGGTA, and RABGGTB) in postmortem dorsolateral prefrontal cortex from patients with schizophrenia and paired comparison subjects (n = 13 pairs). We found decreased levels of FNTA (14%), PGGT1B (13%), and RABGGTB (8%) in schizophrenia. To determine whether upstream or downstream factors may be driving these changes, we also assayed protein expression of the isoprenoid synthases FDPS and GGPS1 and prenylation-dependent processing enzymes RCE and ICMT. We found these upstream and downstream enzymes to have normal protein expression. To rule out effects from chronic antipsychotic treatment, we assayed FNTA, PGGT1B, and RABGGTB in the cortex from rats treated long-term with haloperidol decanoate and found no change in the expression of these proteins. Given the role prenylation plays in localization of key signaling proteins found at the synapse, these data offer a potential mechanism underlying abnormal protein-protein interactions and protein localization in schizophrenia.
Collapse
Affiliation(s)
- Anita L Pinner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA.
| | - Toni M Mueller
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, 43614-2598, USA
| | | | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| |
Collapse
|
29
|
Yang Q, Li B, Tang J, Cui X, Wang Y, Li X, Hu J, Chen Y, Xue W, Lou Y, Qiu Y, Zhu F. Consistent gene signature of schizophrenia identified by a novel feature selection strategy from comprehensive sets of transcriptomic data. Brief Bioinform 2019; 21:1058-1068. [DOI: 10.1093/bib/bbz049] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/11/2019] [Accepted: 03/30/2019] [Indexed: 12/16/2022] Open
Abstract
Abstract
The etiology of schizophrenia (SCZ) is regarded as one of the most fundamental puzzles in current medical research, and its diagnosis is limited by the lack of objective molecular criteria. Although plenty of studies were conducted, SCZ gene signatures identified by these independent studies are found highly inconsistent. As one of the most important factors contributing to this inconsistency, the feature selection methods used currently do not fully consider the reproducibility among the signatures discovered from different datasets. Therefore, it is crucial to develop new bioinformatics tools of novel strategy for ensuring a stable discovery of gene signature for SCZ. In this study, a novel feature selection strategy (1) integrating repeated random sampling with consensus scoring and (2) evaluating the consistency of gene rank among different datasets was constructed. By systematically assessing the identified SCZ signature comprising 135 differentially expressed genes, this newly constructed strategy demonstrated significantly enhanced stability and better differentiating ability compared with the feature selection methods popular in current SCZ research. Based on a first-ever assessment on methods’ reproducibility cross-validated by independent datasets from three representative studies, the new strategy stood out among the popular methods by showing superior stability and differentiating ability. Finally, 2 novel and 17 previously reported transcription factors were identified and showed great potential in revealing the etiology of SCZ. In sum, the SCZ signature identified in this study would provide valuable clues for discovering diagnostic molecules and potential targets for SCZ.
Collapse
Affiliation(s)
- Qingxia Yang
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Bo Li
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Jing Tang
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Xuejiao Cui
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Yunxia Wang
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Xiaofeng Li
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Jie Hu
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuzong Chen
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore, Singapore, Singapore
| | - Weiwei Xue
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yunqing Qiu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing, China
| |
Collapse
|
30
|
Pruski M, Lang B. Primary Cilia-An Underexplored Topic in Major Mental Illness. Front Psychiatry 2019; 10:104. [PMID: 30886591 PMCID: PMC6409319 DOI: 10.3389/fpsyt.2019.00104] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/12/2019] [Indexed: 12/20/2022] Open
Abstract
Though much progress has been made in recent years towards understanding the function and physiology of primary cilia, they remain a somewhat elusive organelle. Some studies have explored the role of primary cilia in the developing nervous system, and their dysfunction has been linked with several neurosensory deficits. Yet, very little has been written on their potential role in psychiatric disorders. This article provides an overview of some of the functions of primary cilia in signalling pathways, and demonstrates that they are a worthy candidate in psychiatric research. The links between primary cilia and major mental illness have been demonstrated to exist at several levels, spanning genetics, signalling pathways, and pharmacology as well as cell division and migration. The primary focus of this review is on the sensory role of the primary cilium and the neurodevelopmental hypothesis of psychiatric disease. As such, the primary cilium is demonstrated to be a key link between the cellular environment and cell behaviour, and hence of key importance in the considerations of the nature and nurture debate in psychiatric research.
Collapse
Affiliation(s)
- Michal Pruski
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Critical Care Laboratory, Critical Care Directorate, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
31
|
Saad MN, Mabrouk MS, Eldeib AM, Shaker OG. Studying the effects of haplotype partitioning methods on the RA-associated genomic results from the North American Rheumatoid Arthritis Consortium (NARAC) dataset. J Adv Res 2019; 18:113-126. [PMID: 30891314 PMCID: PMC6403413 DOI: 10.1016/j.jare.2019.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 12/16/2022] Open
Abstract
Haplotype blocks methods plays a complementary role to the single-SNP approaches. CIT, FGT, SSLD, and single-SNP methods should be applied to discover the markers. Selection of the method used for the association has an impact on the biomarkers. SSLD method detected more significant SNPs than CIT, FGT, and single-SNP methods. The 383 SNPs discovered by all methods are significantly associated with RA.
The human genome, which includes thousands of genes, represents a big data challenge. Rheumatoid arthritis (RA) is a complex autoimmune disease with a genetic basis. Many single-nucleotide polymorphism (SNP) association methods partition a genome into haplotype blocks. The aim of this genome wide association study (GWAS) was to select the most appropriate haplotype block partitioning method for the North American Rheumatoid Arthritis Consortium (NARAC) dataset. The methods used for the NARAC dataset were the individual SNP approach and the following haplotype block methods: the four-gamete test (FGT), confidence interval test (CIT), and solid spine of linkage disequilibrium (SSLD). The measured parameters that reflect the strength of the association between the biomarker and RA were the P-value after Bonferroni correction and other parameters used to compare the output of each haplotype block method. This work presents a comparison among the individual SNP approach and the three haplotype block methods to select the method that can detect all the significant SNPs when applied alone. The GWAS results from the NARAC dataset obtained with the different methods are presented. The individual SNP, CIT, FGT, and SSLD methods detected 541, 1516, 1551, and 1831 RA-associated SNPs respectively, and the individual SNP, FGT, CIT, and SSLD methods detected 65, 156, 159, and 450 significant SNPs respectively, that were not detected by the other methods. Three hundred eighty-three SNPs were discovered by the haplotype block methods and the individual SNP approach, while 1021 SNPs were discovered by all three haplotype block methods. The 383 SNPs detected by all the methods are promising candidates for studying RA susceptibility. A hybrid technique involving all four methods should be applied to detect the significant SNPs associated with RA in the NARAC dataset, but the SSLD method may be preferred because of its advantages when only one method was used.
Collapse
Affiliation(s)
- Mohamed N Saad
- Biomedical Engineering Department, Faculty of Engineering, Minia University, Minia, Egypt
| | - Mai S Mabrouk
- Biomedical Engineering Department, Faculty of Engineering, Misr University for Science and Technology, 6th of October City, Egypt
| | - Ayman M Eldeib
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Giza, Egypt
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
32
|
Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry 2019; 24:549-561. [PMID: 29511299 PMCID: PMC6004314 DOI: 10.1038/s41380-018-0041-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/05/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
Although synaptic loss is thought to be core to the pathophysiology of schizophrenia, the nature, consistency and magnitude of synaptic protein and mRNA changes has not been systematically appraised. Our objective was thus to systematically review and meta-analyse findings. The entire PubMed database was searched for studies from inception date to the 1st of July 2017. We selected case-control postmortem studies in schizophrenia quantifying synaptic protein or mRNA levels in brain tissue. The difference in protein and mRNA levels between cases and controls was extracted and meta-analysis conducted. Among the results, we found a significant reduction in synaptophysin in schizophrenia in the hippocampus (effect size: -0.65, p < 0.01), frontal (effect size: -0.36, p = 0.04), and cingulate cortices (effect size: -0.54, p = 0.02), but no significant changes for synaptophysin in occipital and temporal cortices, and no changes for SNAP-25, PSD-95, VAMP, and syntaxin in frontal cortex. There were insufficient studies for meta-analysis of complexins, synapsins, rab3A and synaptotagmin and mRNA measures. Findings are summarised for these, which generally show reductions in SNAP-25, PSD-95, synapsin and rab3A protein levels in the hippocampus but inconsistency in other regions. Our findings of moderate-large reductions in synaptophysin in hippocampus and frontal cortical regions, and a tendency for reductions in other pre- and postsynaptic proteins in the hippocampus are consistent with models that implicate synaptic loss in schizophrenia. However, they also identify potential differences between regions and proteins, suggesting synaptic loss is not uniform in nature or extent.
Collapse
|
33
|
Datta D, Arnsten AF. Unique Molecular Regulation of Higher-Order Prefrontal Cortical Circuits: Insights into the Neurobiology of Schizophrenia. ACS Chem Neurosci 2018; 9:2127-2145. [PMID: 29470055 DOI: 10.1021/acschemneuro.7b00505] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Schizophrenia is associated with core deficits in cognitive abilities and impaired functioning of the newly evolved prefrontal association cortex (PFC). In particular, neuropathological studies of schizophrenia have found selective atrophy of the pyramidal cell microcircuits in deep layer III of the dorsolateral PFC (dlPFC) and compensatory weakening of related GABAergic interneurons. Studies in monkeys have shown that recurrent excitation in these layer III microcircuits generates the precisely patterned, persistent firing needed for working memory and abstract thought. Importantly, excitatory synapses on layer III spines are uniquely regulated at the molecular level in ways that may render them particularly vulnerable to genetic and/or environmental insults. Glutamate actions are remarkably dependent on cholinergic stimulation, and there are inherent mechanisms to rapidly weaken connectivity, e.g. during stress. In particular, feedforward cyclic adenosine monophosphate (cAMP)-calcium signaling rapidly weakens network connectivity and neuronal firing by opening nearby potassium channels. Many mechanisms that regulate this process are altered in schizophrenia and/or associated with genetic insults. Current data suggest that there are "dual hits" to layer III dlPFC circuits: initial insults to connectivity during the perinatal period due to genetic errors and/or inflammatory insults that predispose the cortex to atrophy, followed by a second wave of cortical loss during adolescence, e.g. driven by stress, at the descent into illness. The unique molecular regulation of layer III circuits may provide a nexus where inflammation disinhibits the neuronal response to stress. Understanding these mechanisms may help to illuminate dlPFC susceptibility in schizophrenia and provide insights for novel therapeutic targets.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, United States
| | - Amy F.T. Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, United States
| |
Collapse
|
34
|
Huang CY, Wang YY, Chen YL, Chen MF, Chiang HS, Kuo PL, Lin YH. CDC42 Negatively Regulates Testis-Specific SEPT12 Polymerization. Int J Mol Sci 2018; 19:ijms19092627. [PMID: 30189608 PMCID: PMC6163814 DOI: 10.3390/ijms19092627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 01/04/2023] Open
Abstract
Septin (SEPT) genes encode well-preserved polymerizing GTP-binding cytoskeletal proteins. The cellular functions of SEPTs consist of mitosis, cytoskeletal remodeling, cell polarity, and vesicle trafficking through interactions with various types of cytoskeletons. We discovered that mutated SEPTIN12 in different codons resulted in teratozoospermia or oligozoospermia. In mouse models with a defective Septin12 allele, sperm morphology was abnormal, sperm count decreased, and sperms were immotile. However, the regulators of SEPT12 are completely unknown. Some studies have indicated that CDC42 negatively regulates the polymerization of SEPT2/6/7 complexes in mammalian cell lines. In this study, we investigated whether CDC42 modulates SEPT12 polymerization and is involved in the terminal differentiation of male germ cells. First, through scanning electron microscopy analysis, we determined that the loss of Septin12 caused defective sperm heads. This indicated that Septin12 is critical for the formation of sperm heads. Second, CDC42 and SEPT12 were similarly localized in the perinuclear regions of the manchette at the head of elongating spermatids, neck region of elongated spermatids, and midpiece of mature spermatozoa. Third, wild-type CDC42 and CDC42Q61L (a constitutive-acting-mutant) substantially repressed SEPT12 polymerization, but CDC42T17N (a dominant-negative-acting mutant) did not, as evident through ectopic expression analysis. We concluded that CDC42 negatively regulates SEPT12 polymerization and is involved in terminal structure formation of sperm heads.
Collapse
Affiliation(s)
- Chia-Yen Huang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan.
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei 106, Taiwan.
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Ya-Yun Wang
- Department of Chemistry, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Ying-Liang Chen
- Department of Environmental Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Mei-Feng Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Han-Sun Chiang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Pao-Lin Kuo
- Department of Obstetrics & Gynecology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| | - Ying-Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| |
Collapse
|
35
|
Gibbons A, Udawela M, Dean B. Non-Coding RNA as Novel Players in the Pathophysiology of Schizophrenia. Noncoding RNA 2018; 4:E11. [PMID: 29657307 PMCID: PMC6027250 DOI: 10.3390/ncrna4020011] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is associated with diverse changes in the brain's transcriptome and proteome. Underlying these changes is the complex dysregulation of gene expression and protein production that varies both spatially across brain regions and temporally with the progression of the illness. The growing body of literature showing changes in non-coding RNA in individuals with schizophrenia offers new insights into the mechanisms causing this dysregulation. A large number of studies have reported that the expression of microRNA (miRNA) is altered in the brains of individuals with schizophrenia. This evidence is complemented by findings that single nucleotide polymorphisms (SNPs) in miRNA host gene sequences can confer an increased risk of developing the disorder. Additionally, recent evidence suggests the expression of other non-coding RNAs, such as small nucleolar RNA and long non-coding RNA, may also be affected in schizophrenia. Understanding how these changes in non-coding RNAs contribute to the development and progression of schizophrenia offers potential avenues for the better treatment and diagnosis of the disorder. This review will focus on the evidence supporting the involvement of non-coding RNA in schizophrenia and its therapeutic potential.
Collapse
Affiliation(s)
- Andrew Gibbons
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
- The Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia.
| | - Madhara Udawela
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
| | - Brian Dean
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
- The Centre for Mental Health, Swinburne University of Technology, Hawthorn, Victoria, Australia.
| |
Collapse
|
36
|
Abstract
Schizophrenia is a complex disorder lacking an effective treatment option for the pervasive and debilitating cognitive impairments experienced by patients. Working memory is a core cognitive function impaired in schizophrenia that depends upon activation of distributed neural network, including the circuitry of the dorsolateral prefrontal cortex (DLPFC). Accordingly, individuals diagnosed with schizophrenia show reduced DLPFC activation while performing working-memory tasks. This lower DLPFC activation appears to be an integral part of the disease pathophysiology, and not simply a reflection of poor performance. Thus, the cellular and circuitry alterations that underlie lower DLPFC neuronal activity in schizophrenia must be determined in order to identify appropriate therapeutic targets. Studies using human postmortem brain tissue provide a robust way to investigate and characterize these cellular and circuitry alterations at multiple levels of resolution, and such studies provide essential information that cannot be obtained either through in vivo studies in humans or through experimental animal models. Studies examining neuronal morphology, protein expression and localization, and transcript levels indicate that a microcircuit composed of excitatory pyramidal cells and inhibitory interneurons containing the calcium-binding protein parvalbumin is altered in the DLPFC of subjects with schizophrenia and likely contributes to DLPFC dysfunction.
Collapse
Affiliation(s)
- Jill R Glausier
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
37
|
Forsyth JK, Lewis DA. Mapping the Consequences of Impaired Synaptic Plasticity in Schizophrenia through Development: An Integrative Model for Diverse Clinical Features. Trends Cogn Sci 2017; 21:760-778. [PMID: 28754595 DOI: 10.1016/j.tics.2017.06.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/13/2017] [Accepted: 06/09/2017] [Indexed: 01/19/2023]
Abstract
Schizophrenia is associated with alterations in sensory, motor, and cognitive functions that emerge before psychosis onset; identifying pathogenic processes that can account for this multi-faceted phenotype remains a challenge. Accumulating evidence suggests that synaptic plasticity is impaired in schizophrenia. Given the role of synaptic plasticity in learning, memory, and neural circuit maturation, impaired plasticity may underlie many features of the schizophrenia syndrome. Here, we summarize the neurobiology of synaptic plasticity, review evidence that plasticity is impaired in schizophrenia, and explore a framework in which impaired synaptic plasticity interacts with brain maturation to yield the emergence of sensory, motor, cognitive, and psychotic features at different times during development in schizophrenia. Key gaps in the literature and future directions for testing this framework are discussed.
Collapse
Affiliation(s)
- Jennifer K Forsyth
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA.
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
38
|
Abstract
Background: Possession of APOEɛ4 is a strong risk factor for late-onset Alzheimer’s disease and is associated with loss of synaptic proteins in the elderly even in the absence of Alzheimer’s disease. Objective: We hypothesized that ɛ4 allele possession in non-demented adults aged under-75 would also be associated with alterations in the levels of synaptic proteins. Methods: We measured synaptophysin, PSD95, drebrin, SNAP-25, and septin 7 by ELISA in hippocampus and superior temporal gyrus from 103 adults aged <75 without dementia. Corresponding gene expression was measured by RT-PCR. Results: There was no evidence that ɛ4 affected levels of the proteins measured. Instead we found an increase in post-synaptic proteins in the hippocampi of those with an ɛ32 genotype. The evidence was strongest for drebrin (p = 0.011). There was some evidence of increased synaptic protein gene expression in ɛ4 carriers. Conclusions: People with an APOEɛ32 genotype have a reduced risk of Alzheimer’s disease. It may be relevant that they have a higher level of post-synaptic proteins in the hippocampus even in earlier adulthood.
Collapse
Affiliation(s)
- Lindsey I Sinclair
- School of Social and Community Medicine, University of Bristol, Oakfield House, Clifton, Bristol, UK
| | - Seth Love
- School of Clinical Sciences, University of Bristol, Level 1 Learning and Research Building, Southmead Hospital, Bristol, UK
| |
Collapse
|
39
|
Synaptic Actin Dysregulation, a Convergent Mechanism of Mental Disorders? J Neurosci 2017; 36:11411-11417. [PMID: 27911743 DOI: 10.1523/jneurosci.2360-16.2016] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/25/2022] Open
Abstract
Actin polymerization governs activity-dependent modulation of excitatory synapses, including their morphology and functionality. It is clear from human genetics that neuropsychiatric and neurodevelopmental disturbances are multigenetic in nature, highlighting the need to better understand the critical neural pathways associated with these disorders and how they are altered by genetic risk alleles. One such signaling pathway that is heavily implicated by candidate genes for psychiatric and neurodevelopmental disorders are regulators of signaling to the actin cytoskeleton, suggesting that its disruption and the ensuring abnormalities of spine structures and postsynaptic complexes is a commonly affected pathway in brain disorders. This review will discuss recent experimental findings that strongly support genetic evidence linking the synaptic cytoskeleton to mental disorders, such as schizophrenia and autism spectrum disorders.
Collapse
|
40
|
Dienel SJ, Bazmi HH, Lewis DA. Development of transcripts regulating dendritic spines in layer 3 pyramidal cells of the monkey prefrontal cortex: Implications for the pathogenesis of schizophrenia. Neurobiol Dis 2017; 105:132-141. [PMID: 28576707 DOI: 10.1016/j.nbd.2017.05.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/22/2017] [Accepted: 05/29/2017] [Indexed: 01/27/2023] Open
Abstract
Certain cognitive deficits in schizophrenia appear to emerge from altered postnatal development of the dorsolateral prefrontal cortex (DLPFC). Dendritic spines on DLPFC layer 3 pyramidal cells are essential for certain cognitive functions, change in density over development, and are reduced in number in schizophrenia. Altered expression of molecular regulators of actin filament assembly and stability, which are essential for spine formation and maintenance, is thought to contribute to the pathogenesis of spine deficits in the disease. However, the normal developmental expression patterns of these molecular regulators of dendritic spines, which might provide insight into the timing of spine deficits in schizophrenia, are unknown. Therefore, we quantified the expression from birth to adulthood of key transcripts regulating dendritic spine density in monkey DLPFC. Layer 3 pyramidal cells, and tissue samples containing layers 3 or 6, were captured by laser microdissection and selected transcripts were quantified using PCR. In layer 3 pyramidal cells, the expression levels of most of the transcripts studied changed early, and not late, in postnatal development. These developmental shifts in expression were generally not detected in tissue homogenates of layers 3 or 6, suggesting that the changes may be enriched in layer 3 pyramidal cells. The timing of these shifts in expression suggests that early, rather than later, postnatal development may be a vulnerable period for layer 3 pyramidal neurons. Disruption of the normal developmental trajectories of these transcripts may contribute to layer 3 pyramidal neuron spine deficits in individuals who are later diagnosed with schizophrenia.
Collapse
Affiliation(s)
- Samuel J Dienel
- Medical Scientist Training Program, University of Pittsburgh, United States; Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States
| | - Holly H Bazmi
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, United States.
| |
Collapse
|
41
|
Layer 3 Excitatory and Inhibitory Circuitry in the Prefrontal Cortex: Developmental Trajectories and Alterations in Schizophrenia. Biol Psychiatry 2017; 81:862-873. [PMID: 27455897 PMCID: PMC5136518 DOI: 10.1016/j.biopsych.2016.05.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 11/20/2022]
Abstract
Convergent evidence suggests that schizophrenia is a disorder of neurodevelopment with alterations in both early and late developmental processes hypothesized to contribute to the disease process. Abnormalities in certain clinical features of schizophrenia, such as working memory impairments, depend on distributed neural circuitry including the dorsolateral prefrontal cortex (DLPFC) and appear to arise during the protracted maturation of this circuitry across childhood and adolescence. In particular, the neural circuitry substrate for working memory in primates involves the coordinated activity of excitatory pyramidal neurons and a specific population of inhibitory gamma-aminobutyric acid neurons (i.e., parvalbumin-containing basket cells) in layer 3 of the DLPFC. Understanding the relationships between the normal development of-and the schizophrenia-associated alterations in-the DLPFC circuitry that subserves working memory could provide new insights into the nature of schizophrenia as a neurodevelopmental disorder. Consequently, we review the following in this article: 1) recent findings regarding alterations of DLPFC layer 3 circuitry in schizophrenia, 2) the developmental refinements in this circuitry that occur during the period when the working memory alterations in schizophrenia appear to arise and progress, and 3) how various adverse environmental exposures could contribute to developmental disturbances of this circuitry in individuals with schizophrenia.
Collapse
|
42
|
Datta D, Arion D, Roman KM, Volk DW, Lewis DA. Altered Expression of ARP2/3 Complex Signaling Pathway Genes in Prefrontal Layer 3 Pyramidal Cells in Schizophrenia. Am J Psychiatry 2017; 174:163-171. [PMID: 27523502 PMCID: PMC5288270 DOI: 10.1176/appi.ajp.2016.16020204] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Lower dendritic spine density on layer 3 pyramidal cells in the dorsolateral prefrontal cortex (DLPFC) appears to contribute to cognitive dysfunction in schizophrenia, whereas psychosis is associated with excessive dopamine release in the striatum. These findings may be related via excitatory projections from the DLPFC to the ventral mesencephalon, the location of dopamine cells projecting to the striatum. Consistent with this hypothesis, deletion of the actin-related protein-2/3 (ARP2/3) complex, which regulates the actin cytoskeleton supporting dendritic spines, produced spine loss in cortical pyramidal cells and striatal hyperdopaminergia in mice. The authors sought to determine whether the ARP2/3 complex is altered in schizophrenia. METHOD In matched pairs of schizophrenia and comparison subjects, transcript levels of ARP2/3 complex signaling pathway were assessed in laser-microdissected DLPFC layer 3 and 5 pyramidal cells and layer 3 parvalbumin interneurons, and in total DLPFC gray matter. RESULTS Transcript levels of ARP2/3 complex subunits and of nucleation promotion factors that regulate the ARP2/3 complex were significantly lower in DLPFC layer 3 and 5 pyramidal cells in schizophrenia. In contrast, these transcripts were unaltered, or only modestly changed, in parvalbumin interneurons and DLPFC gray matter. CONCLUSIONS Down-regulation of the ARP2/3 complex signaling pathway, a common final pathway for multiple signaling cascades that regulate the actin cytoskeleton, would compromise the structural stability of spines, leading to their loss. In concert with findings from deletion of the ARP2/3 complex in mice, these findings support the idea that spine deficits in the DLPFC may contribute to subcortical hyperdopaminergia in schizophrenia.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Translational Neuroscience Program, University of Pittsburgh School of Medicine,Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - Dominique Arion
- Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - Kaitlyn M. Roman
- Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - David W. Volk
- Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| | - David A. Lewis
- Department of Neuroscience, Translational Neuroscience Program, University of Pittsburgh School of Medicine,Department of Neuroscience and Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine
| |
Collapse
|
43
|
Yadav S, Oses-Prieto JA, Peters CJ, Zhou J, Pleasure SJ, Burlingame AL, Jan LY, Jan YN. TAOK2 Kinase Mediates PSD95 Stability and Dendritic Spine Maturation through Septin7 Phosphorylation. Neuron 2017; 93:379-393. [PMID: 28065648 DOI: 10.1016/j.neuron.2016.12.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 10/11/2016] [Accepted: 11/23/2016] [Indexed: 01/09/2023]
Abstract
Abnormalities in dendritic spines are manifestations of several neurodevelopmental and psychiatric diseases. TAOK2 is one of the genes in the 16p11.2 locus, copy number variations of which are associated with autism and schizophrenia. Here, we show that the kinase activity of the serine/threonine kinase encoded by TAOK2 is required for spine maturation. TAOK2 depletion results in unstable dendritic protrusions, mislocalized shaft-synapses, and loss of compartmentalization of NMDA receptor-mediated calcium influx. Using chemical-genetics and mass spectrometry, we identified several TAOK2 phosphorylation targets. We show that TAOK2 directly phosphorylates the cytoskeletal GTPase Septin7, at an evolutionary conserved residue. This phosphorylation induces translocation of Septin7 to the spine, where it associates with and stabilizes the scaffolding protein PSD95, promoting dendritic spine maturation. This study provides a mechanistic basis for postsynaptic stability and compartmentalization via TAOK2-Sept7 signaling, with implications toward understanding the potential role of TAOK2 in neurological deficits associated with the 16p11.2 region.
Collapse
Affiliation(s)
- Smita Yadav
- Departments of Physiology, Biochemistry, and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Juan A Oses-Prieto
- Mass Spectrometry Facility, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christian J Peters
- Departments of Physiology, Biochemistry, and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jing Zhou
- Department of Neurology, Programs in Neuroscience and Developmental Biology, Institute for Regenerative Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Samuel J Pleasure
- Department of Neurology, Programs in Neuroscience and Developmental Biology, Institute for Regenerative Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- Mass Spectrometry Facility, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Y Jan
- Departments of Physiology, Biochemistry, and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh-Nung Jan
- Departments of Physiology, Biochemistry, and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
44
|
Volk DW. Role of microglia disturbances and immune-related marker abnormalities in cortical circuitry dysfunction in schizophrenia. Neurobiol Dis 2016; 99:58-65. [PMID: 28007586 DOI: 10.1016/j.nbd.2016.12.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/14/2016] [Accepted: 12/18/2016] [Indexed: 11/19/2022] Open
Abstract
Studies of genetics, serum cytokines, and autoimmune illnesses suggest that immune-related abnormalities are involved in the disease process of schizophrenia. Furthermore, direct evidence of cortical immune activation, including markedly elevated levels of many immune-related markers, have been reported in the prefrontal cortex in multiple cohorts of schizophrenia subjects. Within the prefrontal cortex in schizophrenia, deficits in the basilar dendritic spines of layer 3 pyramidal neurons and disturbances in inhibitory inputs to pyramidal neurons have also been commonly reported. Interestingly, microglia, the resident immune-related cells of the brain, also regulate excitatory and inhibitory input to pyramidal neurons. Consequently, in this review, we describe the cytological and molecular evidence of immune activation that has been reported in the brains of individuals with schizophrenia and the potential links between these immune-related disturbances with previously reported disturbances in pyramidal and inhibitory neurons in the disorder. Finally, we discuss the role that activated microglia may play in connecting these observations and as potential therapeutic treatment targets in schizophrenia.
Collapse
Affiliation(s)
- David W Volk
- Department of Psychiatry, University of Pittsburgh, W1655 BST, 3811 O'Hara St, Pittsburgh, PA 15213, United States.
| |
Collapse
|
45
|
Chen ML, Tsai FM, Lee MC, Lin YY. Antipsychotic drugs induce cell cytoskeleton reorganization in glial and neuronal cells via Rho/Cdc42 signal pathway. Prog Neuropsychopharmacol Biol Psychiatry 2016; 71:14-26. [PMID: 27302765 DOI: 10.1016/j.pnpbp.2016.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/03/2016] [Accepted: 06/10/2016] [Indexed: 11/24/2022]
Abstract
Long-term administration of antipsychotic drugs (APDs) has been theorized to effect drug-induced changes in protein expression in the brain. Our previous findings revealed that ADPs can regulate Rho GDP-dissociation inhibitor 1 (RhoGDI1) expression in glial cells. To reveal whether APDs (haloperidol, risperidone, and clozapine) might regulate cell functions in rat brain by affecting RhoGDI1, RhoGDI1 regulation, RhoGDI1-related Rho family protein, and also MLC2 in brain of 7-day APD treatment rat were examined. Increased expression of RhoGDI1 and RhoA and decreased expression of MLC2, p-MLC2 and ARP2/3 were found in the cortex of APD-treated rats. The activation of RhoA in APD-treated rat cortex was also found. The regulation of RhoGDI1-induced protein expression and its relation to intracellular stress filament production and cell migration were further examined in APD-treated C6 and B35 cells. APD-induced RhoA expression and activation in C6 cells and Cdc42 expression and activation in B35 cells were investigated. In C6 cells, ARP2/3, ROCK1, pMLC2, and PFN1 expressions were decreased, and N-WASP expression was increased by any of the three APDs. In B35 cells, haloperidol decreased ROCK1 expression, but risperidone increased ROCK1 expression. MLC2, p-MLC2, and PFN1 expressions were decreased in B35 cells treated with either risperidone or clozapine. N-WASP expression was decreased by haloperidol and clozapine. We also found all three APDs enhance C6 and B35 F-actin condensation and migration ability.
Collapse
Affiliation(s)
- Mao-Liang Chen
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC.
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC; Department of Microbiology, Soochow University, Shih Lin, Taipei, Taiwan, ROC
| | - Ming-Cheng Lee
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| | - Yi-Yin Lin
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| |
Collapse
|
46
|
Föcking M, Dicker P, Lopez LM, Hryniewiecka M, Wynne K, English JA, Cagney G, Cotter DR. Proteomic analysis of the postsynaptic density implicates synaptic function and energy pathways in bipolar disorder. Transl Psychiatry 2016; 6:e959. [PMID: 27898073 PMCID: PMC5290351 DOI: 10.1038/tp.2016.224] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022] Open
Abstract
The postsynaptic density (PSD) contains a complex set of proteins of known relevance to neuropsychiatric disorders such as schizophrenia and bipolar disorder. We enriched for this anatomical structure in the anterior cingulate cortex of 16 bipolar disorder samples and 20 controls from the Stanley Medical Research Institute. Unbiased shotgun proteomics incorporating label-free quantitation was used to identify differentially expressed proteins. Quantitative investigation of the PSD identified 2033 proteins, among which 288 were found to be differentially expressed. Validation of expression changes of DNM1, DTNA, NDUFV2, SEPT11 and SSBP was performed by western blotting. Bioinformatics analysis of the differentially expressed proteins implicated metabolic pathways including mitochondrial function, the tricarboxylic acid cycle, oxidative phosphorylation, protein translation and calcium signaling. The data implicate PSD-associated proteins, and specifically mitochondrial function in bipolar disorder. They relate synaptic function in bipolar disorder and the energy pathways that underpin it. Overall, our findings add to a growing literature linking the PSD and mitochondrial function in psychiatric disorders generally, and suggest that mitochondrial function associated with the PSD is particularly important in bipolar disorder.
Collapse
Affiliation(s)
- M Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland,Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Education and Research Centre, Dublin 9, Ireland. E-mail: or
| | - P Dicker
- Departments of Epidemiology and Public Health, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - L M Lopez
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - M Hryniewiecka
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - K Wynne
- Proteome Research Centre, UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - J A English
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - G Cagney
- Proteome Research Centre, UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - D R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland,Department of Psychiatry, Beaumont Hospital, Dublin, Ireland,Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Education and Research Centre, Dublin 9, Ireland. E-mail: or
| |
Collapse
|
47
|
Pinner AL, Tucholski J, Haroutunian V, McCullumsmith RE, Meador-Woodruff JH. Decreased protein S-palmitoylation in dorsolateral prefrontal cortex in schizophrenia. Schizophr Res 2016; 177:78-87. [PMID: 26876311 PMCID: PMC4981568 DOI: 10.1016/j.schres.2016.01.054] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 11/28/2022]
Abstract
Recent reports suggest abnormalities of neurotransmitter receptor trafficking, targeting, dendritic localization, recycling, and degradation in the brain in schizophrenia. We hypothesized that a potential explanation for these findings may be abnormal posttranslational modifications that influence intracellular targeting and trafficking of proteins between subcellular compartments. Dysregulation of protein palmitoylation is a strong candidate for such a process. S-palmitoylation is a reversible thioesterification of palmitoyl-groups to cysteine residues that can regulate trafficking and targeting of intracellular proteins. Using a biotin switch assay to study S-palmitoylation of proteins in human postmortem brain, we identified a pattern of palmitoylated proteins that cluster into 17 bands of discrete molecular masses, including numerous proteins associated with receptor signal transduction. Using mass spectrometry, we identified 219 palmitoylated proteins in human frontal cortex, and individually validated palmitoylation status of a subset of these proteins. Next, we assayed protein palmitoylation in dorsolateral prefrontal cortex from 16 schizophrenia patients and paired comparison subjects. S-palmitoylation was significantly reduced for proteins in most of the 17 schizophrenia bands. In rats chronically treated with haloperidol, the same pattern of palmitoylation was observed but the extent of palmitoylation was unchanged, suggesting that the diminution in protein palmitoylation in schizophrenia is not due to chronic antipsychotic treatment. These results indicate there are changes in the extent of S-palmitoylation of many proteins in the frontal cortex in schizophrenia. Given the roles of this posttranslational modification, these data suggest a potential mechanism reconciling previous observations of abnormal intracellular targeting and trafficking of neurotransmitter receptors in this illness.
Collapse
Affiliation(s)
- Anita L. Pinner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| | - Janusz Tucholski
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, USA
| | - Robert E. McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45219 USA
| | - James H. Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294-0021, USA
| |
Collapse
|
48
|
Rappaz B, Lai Wing Sun K, Correia JP, Wiseman PW, Kennedy TE. FLIM FRET Visualization of Cdc42 Activation by Netrin-1 in Embryonic Spinal Commissural Neuron Growth Cones. PLoS One 2016; 11:e0159405. [PMID: 27482713 PMCID: PMC4970703 DOI: 10.1371/journal.pone.0159405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/02/2016] [Indexed: 12/29/2022] Open
Abstract
Netrin-1 is an essential extracellular chemoattractant that signals through its receptor DCC to guide commissural axon extension in the embryonic spinal cord. DCC directs the organization of F-actin in growth cones by activating an intracellular protein complex that includes the Rho GTPase Cdc42, a critical regulator of cell polarity and directional migration. To address the spatial distribution of signaling events downstream of netrin-1, we expressed the FRET biosensor Raichu-Cdc42 in cultured embryonic rat spinal commissural neurons. Using FLIM-FRET imaging we detected rapid activation of Cdc42 in neuronal growth cones following application of netrin-1. Investigating the signaling mechanisms that control Cdc42 activation by netrin-1, we demonstrate that netrin-1 rapidly enriches DCC at the leading edge of commissural neuron growth cones and that netrin-1 induced activation of Cdc42 in the growth cone is blocked by inhibiting src family kinase signaling. These findings reveal the activation of Cdc42 in embryonic spinal commissural axon growth cones and support the conclusion that src family kinase activation downstream of DCC is required for Cdc42 activation by netrin-1.
Collapse
Affiliation(s)
- Benjamin Rappaz
- Program in NeuroEngineering, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Physics, McGill University, Montreal, QC, H3A 2T8, Canada
| | - Karen Lai Wing Sun
- Program in NeuroEngineering, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - James P. Correia
- Program in NeuroEngineering, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Paul W. Wiseman
- Program in NeuroEngineering, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Physics, McGill University, Montreal, QC, H3A 2T8, Canada
- Department of Chemistry, McGill University, Montreal, QC, H3A 0B8, Canada
| | - Timothy E. Kennedy
- Program in NeuroEngineering, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| |
Collapse
|
49
|
N100 Repetition Suppression Indexes Neuroplastic Defects in Clinical High Risk and Psychotic Youth. Neural Plast 2016; 2016:4209831. [PMID: 26881109 PMCID: PMC4737454 DOI: 10.1155/2016/4209831] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/21/2015] [Accepted: 10/01/2015] [Indexed: 02/06/2023] Open
Abstract
Highly penetrant mutations leading to schizophrenia are enriched for genes coding for N-methyl-D-aspartate receptor signaling complex (NMDAR-SC), implicating plasticity defects in the disease's pathogenesis. The importance of plasticity in neurodevelopment implies a role for therapies that target these mechanisms in early life to prevent schizophrenia. Testing such therapies requires noninvasive methods that can assess engagement of target mechanisms. The auditory N100 is an obligatory cortical response whose amplitude decreases with tone repetition. This adaptation may index the health of plasticity mechanisms required for normal development. We exposed participants aged 5 to 17 years with psychosis (n = 22), at clinical high risk (CHR) for psychosis (n = 29), and healthy controls (n = 17) to an auditory tone repeated 450 times and measured N100 adaptation (mean amplitude during first 150 tones − mean amplitude during last 150 tones). N100 adaptation was reduced in CHR and psychosis, particularly among participants <13 years old. Initial N100 blunting partially accounted for differences. Decreased change in the N100 amplitude with tone repetition may be a useful marker of defects in neuroplastic mechanisms measurable early in life.
Collapse
|
50
|
Lewis DA, Glausier JR. Alterations in Prefrontal Cortical Circuitry and Cognitive Dysfunction in Schizophrenia. NEBRASKA SYMPOSIUM ON MOTIVATION. NEBRASKA SYMPOSIUM ON MOTIVATION 2016; 63:31-75. [PMID: 27627824 DOI: 10.1007/978-3-319-30596-7_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|