1
|
Clarin JD, Bouras NN, Gao WJ. Genetic Diversity in Schizophrenia: Developmental Implications of Ultra-Rare, Protein-Truncating Mutations. Genes (Basel) 2024; 15:1214. [PMID: 39336805 PMCID: PMC11431303 DOI: 10.3390/genes15091214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The genetic basis of schizophrenia (SZ) remains elusive despite its characterization as a highly heritable disorder. This incomplete understanding has led to stagnation in therapeutics and treatment, leaving many suffering with insufficient relief from symptoms. However, recent large-cohort genome- and exome-wide association studies have provided insights into the underlying genetic machinery. The scale of these studies allows for the identification of ultra-rare mutations that confer substantial disease risk, guiding clinicians and researchers toward general classes of genes that are central to SZ etiology. One such large-scale collaboration effort by the Schizophrenia Exome Sequencing Meta-Analysis consortium identified ten, high-risk, ultra-rare, protein-truncating variants, providing the clearest picture to date of the dysfunctional gene products that substantially increase risk for SZ. While genetic studies of SZ provide valuable information regarding "what" genes are linked with the disorder, it is an open question as to "when" during brain development these genetic mutations impose deleterious effects. To shed light on this unresolved aspect of SZ etiology, we queried the BrainSpan developmental mRNA expression database for these ten high-risk genes and discovered three general expression trajectories throughout pre- and postnatal brain development. The elusiveness of SZ etiology, we infer, is not only borne out of the genetic heterogeneity across clinical cases, but also in our incomplete understanding of how genetic mutations perturb neurodevelopment during multiple critical periods. We contextualize this notion within the National Institute of Mental Health's Research Domain Criteria framework and emphasize the utility of considering both genetic variables and developmental context in future studies.
Collapse
Affiliation(s)
- Jacob D Clarin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Nadia N Bouras
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
2
|
Ochoa S, Verdaguer-Rodríguez M, Batlle N, Garreta F, Garcia B, Haro JM, Vila-Andreu È, Hernández MJ, Escandell MJ, Muñoz A, Vilamala S, Marcos S, Bassolas L, Pascua M, Ramos B. Efficacy of the combination of water aerobics and metacognitive training on psychological and physical health variables and their relationship with SP1 and SP4 biomarkers in people with psychosis: a study protocol. Front Psychol 2024; 15:1360004. [PMID: 38919799 PMCID: PMC11197846 DOI: 10.3389/fpsyg.2024.1360004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/16/2024] [Indexed: 06/27/2024] Open
Abstract
Background Metacognitive Training (MCT) is widely used and effective in reducing positive symptoms in psychosis. Physical exercise, such as Water Aerobics (WA), improves general health, quality of life and symptoms as a low impact activity that allows social interactions. Preliminary results suggest a relationship between dopamine and psychotic symptoms, through SP transcription factors, SP1 and SP4 biomarkers. The aims of the project are to evaluate the efficacy of a combined intervention (WA and MCT) for psychosis to improve psychotic symptoms, physical health, and transcription levels of SP biomarkers. Materials and methods This is a unicentric randomized controlled trial of three parallel intervention groups: MCT, WA and combined intervention. The estimated sample will be 48 patients with a psychotic spectrum disorder diagnosis. The assessment will be performed at baseline and at 2-months' follow-up. Instruments used in the assessment will include clinical, cognitive, metacognitive, social cognitive and psychosocial variables. Discussion This will be the first study investigating the impact of the combination of MCT and WA in psychosis. Moreover, it will be the first study analyzing changes in the transcriptional biomarkers SP1 and SP4 after interventions. The results of this study may have clinical implications contributing to the improvement of treatment selection. Clinical trial registration https://clinicaltrials.gov/, identifier: NCT05455593.
Collapse
Affiliation(s)
- Susana Ochoa
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
- Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Marina Verdaguer-Rodríguez
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
- Clinical and Health Psychology Department, Psychology Faculty, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Núria Batlle
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
| | | | | | - Josep María Haro
- Parc Sanitari Sant Joan de Déu, España, Spain
- Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
- Epidemiologia dels trastorns mentals i de l'envelliment, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Èlia Vila-Andreu
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
| | | | - Maria José Escandell
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
| | - Ana Muñoz
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
| | - Sònia Vilamala
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
| | | | | | | | - Belén Ramos
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
- Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
- Facultat de Medicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia, Vic, Spain
| | - on behalf of Thalassa Research Group
- Etiopatogènia i Tractament dels Trastorns Mentals Greus (MERITT), Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Parc Sanitari Sant Joan de Déu, España, Spain
| |
Collapse
|
3
|
Schumacher MA. Peripheral Neuroinflammation and Pain: How Acute Pain Becomes Chronic. Curr Neuropharmacol 2024; 22:6-14. [PMID: 37559537 PMCID: PMC10716877 DOI: 10.2174/1570159x21666230808111908] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/05/2023] [Accepted: 04/26/2023] [Indexed: 08/11/2023] Open
Abstract
The number of individuals suffering from severe chronic pain and its social and financial impact is staggering. Without significant advances in our understanding of how acute pain becomes chronic, effective treatments will remain out of reach. This mini review will briefly summarize how critical signaling pathways initiated during the early phases of peripheral nervous system inflammation/ neuroinflammation establish long-term modifications of sensory neuronal function. Together with the recruitment of non-neuronal cellular elements, nociceptive transduction is transformed into a pathophysiologic state sustaining chronic peripheral sensitization and pain. Inflammatory mediators, such as nerve growth factor (NGF), can lower activation thresholds of sensory neurons through posttranslational modification of the pain-transducing ion channels transient-receptor potential TRPV1 and TRPA1. Performing a dual role, NGF also drives increased expression of TRPV1 in sensory neurons through the recruitment of transcription factor Sp4. More broadly, Sp4 appears to modulate a nociceptive transcriptome including TRPA1 and other genes encoding components of pain transduction. Together, these findings suggest a model where acute pain evoked by peripheral injury-induced inflammation becomes persistent through repeated cycles of TRP channel modification, Sp4-dependent overexpression of TRP channels and ongoing production of inflammatory mediators.
Collapse
Affiliation(s)
- Mark A Schumacher
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, California, 94143 USA
| |
Collapse
|
4
|
Young JW. Development of cross-species translational paradigms for psychiatric research in the Research Domain Criteria era. Neurosci Biobehav Rev 2023; 148:105119. [PMID: 36889561 DOI: 10.1016/j.neubiorev.2023.105119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
The past 30 years of IBNS has included research attempting to treat the cognitive and behavioral deficits observed in people with psychiatric conditions. Early work utilized drugs identified from tests thought to be cognition-relevant, however the high failure rate crossing the translational-species barrier led to focus on developing valid cross-species translational tests. The face, predictive, and neurobiological validities used to assess animal models of psychiatry can be used to validate these tests. Clinical sensitivity is another important aspect however, for if the clinical population targeted for treatment does not exhibit task deficits, then why develop treatments? This review covers some work validating cross-species translational tests and suggests future directions. Also covered is the contribution IBNS made to fostering such research and my role in IBNS, making it more available to all including fostering mentor/mentee programs plus spearheading diversity and inclusivity initiatives. All science needs support and IBNS has supported research recreating the behavioral abnormalities that define psychiatric conditions with the aim to improve the lives of people with such conditions.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
5
|
Xu H, Jia J, Jeong HH, Zhao Z. Deep learning for detecting and elucidating human T-cell leukemia virus type 1 integration in the human genome. PATTERNS (NEW YORK, N.Y.) 2023; 4:100674. [PMID: 36873907 PMCID: PMC9982299 DOI: 10.1016/j.patter.2022.100674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 12/13/2022] [Indexed: 02/12/2023]
Abstract
Human T-cell leukemia virus type 1 (HTLV-1), a retrovirus, is the causative agent for adult T cell leukemia/lymphoma and many other human diseases. Accurate and high throughput detection of HTLV-1 virus integration sites (VISs) across the host genomes plays a crucial role in the prevention and treatment of HTLV-1-associated diseases. Here, we developed DeepHTLV, the first deep learning framework for VIS prediction de novo from genome sequence, motif discovery, and cis-regulatory factor identification. We demonstrated the high accuracy of DeepHTLV with more efficient and interpretive feature representations. Decoding the informative features captured by DeepHTLV resulted in eight representative clusters with consensus motifs for potential HTLV-1 integration. Furthermore, DeepHTLV revealed interesting cis-regulatory elements in regulation of VISs that have significant association with the detected motifs. Literature evidence demonstrated nearly half (34) of the predicted transcription factors enriched with VISs were involved in HTLV-1-associated diseases. DeepHTLV is freely available at https://github.com/bsml320/DeepHTLV.
Collapse
Affiliation(s)
- Haodong Xu
- Center for Precision Health, School of Biomedical Informatics, UTHealth Science Center at Houston, Houston, TX 77030, USA
| | - Johnathan Jia
- Center for Precision Health, School of Biomedical Informatics, UTHealth Science Center at Houston, Houston, TX 77030, USA.,MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Hyun-Hwan Jeong
- Center for Precision Health, School of Biomedical Informatics, UTHealth Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, UTHealth Science Center at Houston, Houston, TX 77030, USA.,MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.,Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| |
Collapse
|
6
|
Shahcheraghi SH, Ayatollahi J, Lotfi M, Aljabali AAA, Al-Zoubi MS, Panda PK, Mishra V, Satija S, Charbe NB, Serrano-Aroca Á, Bahar B, Takayama K, Goyal R, Bhatia A, Almutary AG, Alnuqaydan AM, Mishra Y, Negi P, Courtney A, McCarron PA, Bakshi HA, Tambuwala MM. Gene Therapy for Neuropsychiatric Disorders: Potential Targets and Tools. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:51-65. [PMID: 35249508 DOI: 10.2174/1871527321666220304153719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/16/2022] [Accepted: 01/16/2022] [Indexed: 01/01/2023]
Abstract
Neuropsychiatric disorders that affect the central nervous system cause considerable pressures on the health care system and have a substantial economic burden on modern societies. The present treatments based on available drugs are mostly ineffective and often costly. The molecular process of neuropsychiatric disorders is closely connected to modifying the genetic structures inherited or caused by damage, toxic chemicals, and some current diseases. Gene therapy is presently an experimental concept for neurological disorders. Clinical applications endeavor to alleviate the symptoms, reduce disease progression, and repair defective genes. Implementing gene therapy in inherited and acquired neurological illnesses entails the integration of several scientific disciplines, including virology, neurology, neurosurgery, molecular genetics, and immunology. Genetic manipulation has the power to minimize or cure illness by inducing genetic alterations at endogenous loci. Gene therapy that involves treating the disease by deleting, silencing, or editing defective genes and delivering genetic material to produce therapeutic molecules has excellent potential as a novel approach for treating neuropsychiatric disorders. With the recent advances in gene selection and vector design quality in targeted treatments, gene therapy could be an effective approach. This review article will investigate and report the newest and the most critical molecules and factors in neuropsychiatric disorder gene therapy. Different genome editing techniques available will be evaluated, and the review will highlight preclinical research of genome editing for neuropsychiatric disorders while also evaluating current limitations and potential strategies to overcome genome editing advancements.
Collapse
Affiliation(s)
- Seyed H Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Jamshid Ayatollahi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alaa A A Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Mazhar S Al-Zoubi
- Yarmouk University, Faculty of Medicine, Department of Basic Medical Sciences, Irbid, Jordan
| | - Pritam K Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, 75120 Uppsala, Sweden
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Nitin B Charbe
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Translational Research Centre San Alberto Magno, Catholic University of Valencia San Vicente Mártir, C/Guillem de Castro 94, 46001 Valencia, Spain
| | - Bojlul Bahar
- Nutrition Sciences and Applied Food Safety Studies, Research Centre for Global Development, School of Sport & Health Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Kazuo Takayama
- Center for IPS Cell Research and Application, Kyoto University, Kyoto, 606-8397, Japan
| | - Rohit Goyal
- Neuropharmacology Laboratory, School of Pharmaceutical Sciences, Shoolini University, Post Box No. 9, Solan, Himachal Pradesh 173212, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Punjab 151001, India
| | - Abdulmajeed G Almutary
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Saudi Arabia
| | - Abdullah M Alnuqaydan
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Saudi Arabia
| | - Yachana Mishra
- Shri Shakti Degree College, Sankhahari, Ghatampur 209206, India
| | - Poonam Negi
- Shoolini University of Biotechnology and Management Sciences, Solan 173 212, India
| | - Aaron Courtney
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Paul A McCarron
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Hamid A Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| |
Collapse
|
7
|
Analysis of Molecular Networks in the Cerebellum in Chronic Schizophrenia: Modulation by Early Postnatal Life Stressors in Murine Models. Int J Mol Sci 2021; 22:ijms221810076. [PMID: 34576238 PMCID: PMC8469990 DOI: 10.3390/ijms221810076] [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: 05/31/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 01/17/2023] Open
Abstract
Despite the growing importance of the cerebellum as a region highly vulnerable to accumulating molecular errors in schizophrenia, limited information is available regarding altered molecular networks with potential therapeutic targets. To identify altered networks, we conducted one-shot liquid chromatography–tandem mass spectrometry in postmortem cerebellar cortex in schizophrenia and healthy individuals followed by bioinformatic analysis (PXD024937 identifier in ProteomeXchange repository). A total of 108 up-regulated proteins were enriched in stress-related proteins, half of which were also enriched in axonal cytoskeletal organization and vesicle-mediated transport. A total of 142 down-regulated proteins showed an enrichment in proteins involved in mitochondrial disease, most of which were also enriched in energy-related biological functions. Network analysis identified a mixed module of mainly axonal-related pathways for up-regulated proteins with a high number of interactions for stress-related proteins. Energy metabolism and neutrophil degranulation modules were found for down-regulated proteins. Further, two double-hit postnatal stress murine models based on maternal deprivation combined with social isolation or chronic restraint stress were used to investigate the most robust candidates of generated networks. CLASP1 from the axonal module in the model of maternal deprivation was combined with social isolation, while YWHAZ was not altered in either model. METTL7A from the degranulation pathway was reduced in both models and was identified as altered also in previous gene expression studies, while NDUFB9 from the energy network was reduced only in the model of maternal deprivation combined with social isolation. This work provides altered stress- and mitochondrial disease-related proteins involved in energy, immune and axonal networks in the cerebellum in schizophrenia as possible novel targets for therapeutic interventions and suggests that METTL7A is a possible relevant altered stress-related protein in this context.
Collapse
|
8
|
The Potential Use of Peripheral Blood Mononuclear Cells as Biomarkers for Treatment Response and Outcome Prediction in Psychiatry: A Systematic Review. Mol Diagn Ther 2021; 25:283-299. [PMID: 33978935 DOI: 10.1007/s40291-021-00516-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Psychiatric disorders have a major impact on the global burden of disease while therapeutic interventions remain insufficient to adequately treat a large number of patients. Regrettably, the efficacy of several psychopharmacological treatment regimens becomes apparent only after 4-6 weeks, and at this point, a significant number of patients present as non-responsive. As such, many patients go weeks/months without appropriate treatment or symptom management. Adequate biomarkers for treatment success and outcome prediction are thus urgently needed. OBJECTIVE With this systematic review, we provide an overview of the use of peripheral blood mononuclear cells (PBMCs) and their signaling pathways in evaluating and/or predicting the effectiveness of different treatment regimens in the course of psychiatric illnesses. We highlight PBMC characteristics that (i) reflect treatment presence, (ii) allow differentiation of responders from non-responders, and (iii) prove predictive at baseline with regard to treatment outcome for a broad range of psychiatric intervention strategies. REVIEW METHODS A PubMed database search was performed to extract papers investigating the relation between any type of PBMC characteristic and treatment presence and/or outcome in patients suffering from severe mental illness. Criteria for eligibility were: written in English; psychiatric diagnosis based on DSM-III-R or newer; PBMC isolation via gradient centrifugation; comparison between treated and untreated patients via PBMC features; sample size ≥ n = 5 per experimental group. Papers not researching in vivo treatment effects between patients and healthy controls, non-clinical trials, and non-hypothesis-/data-driven (e.g., -omics designs) approaches were excluded. DATA SYNTHESIS Twenty-nine original articles were included and qualitatively summarized. Antidepressant and antipsychotic treatments were mostly reflected by intracellular inflammatory markers while intervention with mood stabilizers was evidenced through cell maturation pathways. Lastly, cell viability parameters mirrored predominantly non-pharmacological therapeutic strategies. As for response prediction, PBMC (subtype) counts and telomerase activity seemed most promising for antidepressant treatment outcome determination; full length brain-derived neurotrophic factor (BDNF)/truncated BDNF were shown to be most apt to prognosticate antipsychotic treatment. CONCLUSIONS We conclude that, although inherent limitations to and heterogeneity in study designs in combination with the scarce number of original studies hamper unambiguous identification, several PBMC characteristics-mostly related to inflammatory pathways and cell viability-indeed show promise towards establishment as clinically relevant treatment biomarkers.
Collapse
|
9
|
Arasappan D, Eickhoff SB, Nemeroff CB, Hofmann HA, Jabbi M. Transcription Factor Motifs Associated with Anterior Insula Gene Expression Underlying Mood Disorder Phenotypes. Mol Neurobiol 2021; 58:1978-1989. [PMID: 33411239 DOI: 10.1007/s12035-020-02195-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/30/2020] [Indexed: 10/22/2022]
Abstract
Mood disorders represent a major cause of morbidity and mortality worldwide but the brain-related molecular pathophysiology in mood disorders remains largely undefined. Because the anterior insula is reduced in volume in patients with mood disorders, RNA was extracted from the anterior insula postmortem anterior insula of mood disorder samples and compared with unaffected controls for RNA-sequencing identification of differentially expressed genes (DEGs) in (a) bipolar disorder (BD; n = 37) versus (vs.) controls (n = 33), and (b) major depressive disorder (MDD n = 30) vs. controls, and (c) low vs. high axis I comorbidity (a measure of cumulative psychiatric disease burden). Given the regulatory role of transcription factors (TFs) in gene expression via specific-DNA-binding domains (motifs), we used JASPAR TF binding database to identify TF-motifs. We found that DEGs in BD vs. controls, MDD vs. controls, and high vs. low axis I comorbidity were associated with TF-motifs that are known to regulate expression of toll-like receptor genes, cellular homeostatic-control genes, and genes involved in embryonic, cellular/organ, and brain development. Robust imaging-guided transcriptomics by using meta-analytic imaging results to guide independent postmortem dissection for RNA-sequencing was applied by targeting the gray matter volume reduction in the anterior insula in mood disorders, to guide independent postmortem identification of TF motifs regulating DEG. Our findings of TF-motifs that regulate the expression of immune, cellular homeostatic-control, and developmental genes provide novel information about the hierarchical relationship between gene regulatory networks, the TFs that control them, and proximate underlying neuroanatomical phenotypes in mood disorders.
Collapse
Affiliation(s)
- Dhivya Arasappan
- Center for Biomedical Research Support, University of Texas at Austin, Austin, TX, USA
| | - Simon B Eickhoff
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany
| | - Charles B Nemeroff
- Department of Psychiatry, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- The Mulva Clinic for Neurosciences, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Institute of Early Life Adversity Research, Austin, TX, USA
| | - Hans A Hofmann
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - Mbemba Jabbi
- Department of Psychiatry, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- The Mulva Clinic for Neurosciences, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA.
- Department of Psychology, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
10
|
Serwach K, Gruszczynska-Biegala J. Target Molecules of STIM Proteins in the Central Nervous System. Front Mol Neurosci 2020; 13:617422. [PMID: 33424550 PMCID: PMC7786003 DOI: 10.3389/fnmol.2020.617422] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/02/2020] [Indexed: 12/16/2022] Open
Abstract
Stromal interaction molecules (STIMs), including STIM1 and STIM2, are single-pass transmembrane proteins that are located predominantly in the endoplasmic reticulum (ER). They serve as calcium ion (Ca2+) sensors within the ER. In the central nervous system (CNS), they are involved mainly in Orai-mediated store-operated Ca2+ entry (SOCE). The key molecular components of the SOCE pathway are well-characterized, but the molecular mechanisms that underlie the regulation of this pathway need further investigation. Numerous intracellular target proteins that are located in the plasma membrane, ER, cytoskeleton, and cytoplasm have been reported to play essential roles in concert with STIMs, such as conformational changes in STIMs, their translocation, the stabilization of their interactions with Orai, and the activation of other channels. The present review focuses on numerous regulators, such as Homer, SOCE-associated regulatory factor (SARAF), septin, synaptopodin, golli proteins, partner of STIM1 (POST), and transcription factors and proteasome inhibitors that regulate STIM-Orai interactions in the CNS. Further we describe novel roles of STIMs in mediating Ca2+ influx via other than Orai pathways, including TRPC channels, VGCCs, AMPA and NMDA receptors, and group I metabotropic glutamate receptors. This review also summarizes recent findings on additional molecular targets of STIM proteins including SERCA, IP3Rs, end-binding proteins (EB), presenilin, and CaMKII. Dysregulation of the SOCE-associated toolkit, including STIMs, contributes to the development of neurodegenerative disorders (e.g., Alzheimer's disease, Parkinson's disease, and Huntington's disease), traumatic brain injury, epilepsy, and stroke. Emerging evidence points to the role of STIM proteins and several of their molecular effectors and regulators in neuronal and glial physiology and pathology, suggesting their potential application for future therapeutic strategies.
Collapse
Affiliation(s)
- Karolina Serwach
- Molecular Biology Unit, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
11
|
Lelkes Z. Ouabain, a Na–K-ATPase inhibitor, enhances wakefulness in rats. Neuropharmacology 2020; 176:108224. [DOI: 10.1016/j.neuropharm.2020.108224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 10/23/2022]
|
12
|
Vidal-Domènech F, Riquelme G, Pinacho R, Rodriguez-Mias R, Vera A, Monje A, Ferrer I, Callado LF, Meana JJ, Villén J, Ramos B. Calcium-binding proteins are altered in the cerebellum in schizophrenia. PLoS One 2020; 15:e0230400. [PMID: 32639965 PMCID: PMC7343173 DOI: 10.1371/journal.pone.0230400] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
Alterations in the cortico-cerebellar-thalamic-cortical circuit might underlie the diversity of symptoms in schizophrenia. However, molecular changes in cerebellar neuronal circuits, part of this network, have not yet been fully determined. Using LC-MS/MS, we screened altered candidates in pooled grey matter of cerebellum from schizophrenia subjects who committed suicide (n = 4) and healthy individuals (n = 4). Further validation by immunoblotting of three selected candidates was performed in two cohorts comprising schizophrenia (n = 20), non-schizophrenia suicide (n = 6) and healthy controls (n = 21). We found 99 significantly altered proteins, 31 of them previously reported in other brain areas by proteomic studies. Transport function was the most enriched category, while cell communication was the most prevalent function. For validation, we selected the vacuolar proton pump subunit 1 (VPP1), from transport, and two EF-hand calcium-binding proteins, calmodulin and parvalbumin, from cell communication. All candidates showed significant changes in schizophrenia (n = 7) compared to controls (n = 7). VPP1 was altered in the non-schizophrenia suicide group and increased levels of parvalbumin were linked to antipsychotics. Further validation in an independent cohort of non-suicidal chronic schizophrenia subjects (n = 13) and non-psychiatric controls (n = 14) showed that parvalbumin was increased, while calmodulin was decreased in schizophrenia. Our findings provide evidence of calcium-binding protein dysregulation in the cerebellum in schizophrenia, suggesting an impact on normal calcium-dependent synaptic functioning of cerebellar circuits. Our study also links VPP1 to suicide behaviours, suggesting a possible impairment in vesicle neurotransmitter refilling and release in these phenotypes.
Collapse
Affiliation(s)
- Francisco Vidal-Domènech
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Dept. de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Gemma Riquelme
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Raquel Pinacho
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Ricard Rodriguez-Mias
- Department of Genome Sciences, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - América Vera
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Alfonso Monje
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Spain
| | - Isidre Ferrer
- Departamento de Patologia y Terapeutica Experimental, Universidad de Barcelona, Senior consultant Servicio Anatomia Patológica, Hospital Universitario de Bellvitge-IDIBELL, CIBERNED, Hospital de Llobregat, Barcelona, Spain
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, CIBERSAM, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - J. Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, CIBERSAM, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Judit Villén
- Department of Genome Sciences, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Belén Ramos
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Dept. de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, CIBERSAM, Spain
- * E-mail:
| |
Collapse
|
13
|
Saucedo‐Uribe E, Genis‐Mendoza AD, Díaz‐Anzaldúa A, Martínez‐Magaña JJ, Tovilla‐Zarate CA, Juárez‐Rojop I, Lanzagorta N, Escamilla M, González‐Castro TB, López Narvaez ML, Hernández‐Díaz Y, Nicolini H. Differential effects on neurodevelopment of FTO variants in obesity and bipolar disorder suggested by in silico prediction of functional impact: An analysis in Mexican population. Brain Behav 2019; 9:e01249. [PMID: 31033179 PMCID: PMC6576176 DOI: 10.1002/brb3.1249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/08/2019] [Accepted: 02/10/2019] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Several studies indicate that polygenic obesity is linked to fat-mass and obesity-associated (FTO) genetic variants. Nevertheless, the link between variants in FTO and mental disorders has been barely explored. The present work aims to determine whether FTO genetic variants are associated with bipolar disorder and obesity, and to perform an in silico prediction of variant-dependent functional impact on the developing brain transcriptome. METHODS Four hundred and forty-six Mexican mestizos were included in a genetic association analysis. SNP-sequence kernel association test and linear mixed models were implemented for genetic association assessment. For functional impact prediction, we analyzed the mapping of regulatory elements, the modification of binding sites of transcription factors and the expression of transcription factors in the brain developing transcriptome, searching on different databases. RESULTS In the set-based analysis, we found different associated regions to BD (bipolar disorder) and obesity. The promoter flanking region of FTO intron 1 was associated with differential effects on BMI, while intron 2 of RPGRIP1L and FTO upstream regions were associated with BD. The prediction analysis showed that FTO BD-associated variants disturb binding sites of SP1 and SP2; obesity-associated variants, on the other hand, disturb binding sites of FOXP1, which are transcription factors highly expressed during prenatal development stages of the brain. CONCLUSION Our results suggest a possible effect of FTO variants on neurodevelopment in obesity and bipolar disorder, which gives new insights into the molecular mechanism underlying this association.
Collapse
Affiliation(s)
- Erasmo Saucedo‐Uribe
- Center of Advanced NeurosciencesDepartment of PsychiatryAutonomous University of Nuevo LeonHospital Universitario “Dr. José Eleuterio González”MonterreyMexico
| | - Alma Delia Genis‐Mendoza
- Laboratory of Genomics of Psychiatric and Neurodegenerative DiseasesNational Institute of Genomic MedicineMexico CityMexico
- Children's Psychiatric Hospital “Dr. Juan N. Navarro”Psychiatric Attention ServicesMexico CityMexico
| | - Adriana Díaz‐Anzaldúa
- Department of Psychiatric GeneticsClinical InvestigationsNational Institute of Psychiatry Ramón de la Fuente MuñizMexico CityMexico
| | - José Jaime Martínez‐Magaña
- Laboratory of Genomics of Psychiatric and Neurodegenerative DiseasesNational Institute of Genomic MedicineMexico CityMexico
| | | | - Isela Juárez‐Rojop
- Academic Division of Health SciencesAutonomous University of TabascoVillahermosaTabascoMexico
| | - Nuria Lanzagorta
- Department of Clinical ResearchCarracci Medical GroupMexico CityMexico
| | - Michael Escamilla
- Center of Emphasis in NeurosciencesHealth Sciences CenterTexas Tech UniversityEl Paso, TexasUSA
| | | | | | - Yazmín Hernández‐Díaz
- Multidisciplinary Academic Division of Jalpa de MendezUniversidad Juárez Autónoma de TabascoComalcalcoTabascoMexico
| | - Humberto Nicolini
- Laboratory of Genomics of Psychiatric and Neurodegenerative DiseasesNational Institute of Genomic MedicineMexico CityMexico
- Department of Clinical ResearchCarracci Medical GroupMexico CityMexico
| |
Collapse
|
14
|
Sheehan K, Lee J, Chong J, Zavala K, Sharma M, Philipsen S, Maruyama T, Xu Z, Guan Z, Eilers H, Kawamata T, Schumacher M. Transcription factor Sp4 is required for hyperalgesic state persistence. PLoS One 2019; 14:e0211349. [PMID: 30811405 PMCID: PMC6392229 DOI: 10.1371/journal.pone.0211349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022] Open
Abstract
Understanding how painful hypersensitive states develop and persist beyond the initial hours to days is critically important in the effort to devise strategies to prevent and/or reverse chronic painful states. Changes in nociceptor transcription can alter the abundance of nociceptive signaling elements, resulting in longer-term change in nociceptor phenotype. As a result, sensitized nociceptive signaling can be further amplified and nocifensive behaviors sustained for weeks to months. Building on our previous finding that transcription factor Sp4 positively regulates the expression of the pain transducing channel TRPV1 in Dorsal Root Ganglion (DRG) neurons, we sought to determine if Sp4 serves a broader role in the development and persistence of hypersensitive states in mice. We observed that more than 90% of Sp4 staining DRG neurons were small to medium sized, primarily unmyelinated (NF200 neg) and the majority co-expressed nociceptor markers TRPV1 and/or isolectin B4 (IB4). Genetically modified mice (Sp4+/-) with a 50% reduction of Sp4 showed a reduction in DRG TRPV1 mRNA and neuronal responses to the TRPV1 agonist-capsaicin. Importantly, Sp4+/- mice failed to develop persistent inflammatory thermal hyperalgesia, showing a reversal to control values after 6 hours. Despite a reversal of inflammatory thermal hyperalgesia, there was no difference in CFA-induced hindpaw swelling between CFA Sp4+/- and CFA wild type mice. Similarly, Sp4+/- mice failed to develop persistent mechanical hypersensitivity to hind-paw injection of NGF. Although Sp4+/- mice developed hypersensitivity to traumatic nerve injury, Sp4+/- mice failed to develop persistent cold or mechanical hypersensitivity to the platinum-based chemotherapeutic agent oxaliplatin, a non-traumatic model of neuropathic pain. Overall, Sp4+/- mice displayed a remarkable ability to reverse the development of multiple models of persistent inflammatory and neuropathic hypersensitivity. This suggests that Sp4 functions as a critical control point for a network of genes that conspire in the persistence of painful hypersensitive states.
Collapse
Affiliation(s)
- Kayla Sheehan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Jessica Lee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Jillian Chong
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Kathryn Zavala
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Manohar Sharma
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tomoyuki Maruyama
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Zheyun Xu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Helge Eilers
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Tomoyuki Kawamata
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
Salagre E, Arango C, Artigas F, Ayuso-Mateos JL, Bernardo M, Castro-Fornieles J, Bobes J, Desco M, Fañanás L, González-Pinto A, Haro JM, Leza JC, Mckenna PJ, Meana JJ, Menchón JM, Micó JA, Palomo T, Pazos Á, Pérez V, Saiz-Ruiz J, Sanjuán J, Tabarés-Seisdedos R, Crespo-Facorro B, Casas M, Vilella E, Palao D, Olivares JM, Rodriguez-Jimenez R, Vieta E. CIBERSAM: Ten years of collaborative translational research in mental disorders. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2018; 12:1-8. [PMID: 30416047 DOI: 10.1016/j.rpsm.2018.10.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Estela Salagre
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servicio de Psiquiatría y Psicología, Hospital Clínic, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España
| | - Celso Arango
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servicio de Psiquiatría del Niño y del Adolescente, Hospital General Universitario Gregorio Marañón (IiSGM), Facultad de Medicina, Universidad Complutense, CIBERSAM, Madrid, España
| | - Francesc Artigas
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Neuroquímica y Neurofarmacología, Institut d'Investigacions Biomèdiques de Barcelona IIBB-CSIC, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España
| | - José Luis Ayuso-Mateos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Psiquiatría, Universidad Autónoma de Madrid, Madrid, España
| | - Miquel Bernardo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Unidad Esquizofrenia Clínic, Institut Clínic de Neurociencias, Hospital Clínic, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España
| | - Josefina Castro-Fornieles
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servicio de Psiquiatría y Psicología Infantil y Juvenil, Institut Clínic de Neurociencias, IDIBAPS, Hospital Clínic de Barcelona, Barcelona, España
| | - Julio Bobes
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Área de Psiquiatría, Universidad de Oviedo, Servicio de Salud del Principado de Asturias, Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Asturias, España
| | - Manuel Desco
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Instituto de Investigación Sanitaria Gregorio Marañón, Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, España
| | - Lourdes Fañanás
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Secció Zoologia i Antropologia Biològica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, España
| | - Ana González-Pinto
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Psiquiatría, Hospital Universitario Araba, Instituto de Investigación Sanitaria Bioaraba; Universidad del País Vasco, Vitoria, España
| | - Josep María Haro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Sant Boi de Llobregat, Barcelona, España
| | - Juan Carlos Leza
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto Universitario de Investigación en Neuroquímica UCM, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, España
| | - Peter J Mckenna
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; FIDMAG Germanes Hospitalàries Research Foundation, Sant Boi de Llobregat, Barcelona, España
| | - José Javier Meana
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Bizkaia, Instituto de Investigación Sanitaria Biocruces Bizkaia, Barakaldo, Barakaldo, Bizkaia, España
| | - José Manuel Menchón
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servicio de Psiquiatría, Hospital Universitari de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat; Departamento de Ciencias Clínicas, Universitat de Barcelona, Barcelona, España
| | - Juan Antonio Micó
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Grupo de Investigación en Neuropsicofarmacología y Psicobiología,, Departamento de Neurociencias, Universidad de Cádiz, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, España
| | - Tomás Palomo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Psiquiatría, Hospital 12 de Octubre, Madrid, España
| | - Ángel Pazos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Cantabria, España
| | - Víctor Pérez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Institut de Neuropsiquiatria i Addiccions, Hospital del Mar, Universitat Autònoma de Barcelona, Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, España
| | - Jerónimo Saiz-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Psiquiatría, Hospital Ramon y Cajal, Universidad de Alcalá, IRYCIS, Madrid, España
| | - Julio Sanjuán
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; INCLIVA, Universidad de Valencia, Hospital Clínico Universitario de Valencia, Valencia, España
| | - Rafael Tabarés-Seisdedos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Medicina, INCLIVA, Universidad de Valencia, Valencia, España
| | - Benedicto Crespo-Facorro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Medicina y Psiquiatría, Universidad de Cantabria, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Cantabria, España
| | - Miquel Casas
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servicio de Psiquiatría, Hospital Universitari Vall d'Hebron, Departamento de Psiquiatría y Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, España
| | - Elisabet Vilella
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Tarragona, España
| | - Diego Palao
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servei de Salut Mental, Parc Taulí Hospital Universitari, Institut de Recerca i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Barcelona, España
| | - Jose Manuel Olivares
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Unidad de Psiquiatría, Hospital Álvaro Cunqueiro, Complejo Hospitalario Universitario de Vigo, Instituto Biomédico Galicia Sur, Vigo, Pontevedra, España
| | - Roberto Rodriguez-Jimenez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Departamento de Psiquiatría, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), CogPsy-Group, Universidad Complutense de Madrid (UCM), Madrid, España
| | - Eduard Vieta
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, España; Servicio de Psiquiatría y Psicología, Hospital Clínic, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España.
| |
Collapse
|
16
|
Wang X, Pinto-Duarte A, Behrens MM, Zhou X, Sejnowski TJ. Ketamine independently modulated power and phase-coupling of theta oscillations in Sp4 hypomorphic mice. PLoS One 2018. [PMID: 29513708 PMCID: PMC5841791 DOI: 10.1371/journal.pone.0193446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Reduced expression of Sp4, the murine homolog of human SP4, a risk gene of multiple psychiatric disorders, led to N-methyl-D-aspartate (NMDA) hypofunction in mice, producing behavioral phenotypes reminiscent of schizophrenia, including hypersensitivity to ketamine. As accumulating evidence on molecular mechanisms and behavioral phenotypes established Sp4 hypomorphism as a promising animal model, systems-level neural circuit mechanisms of Sp4 hypomorphism, especially network dynamics underlying cognitive functions, remain poorly understood. We attempted to close this gap in knowledge in the present study by recording multi-channel epidural electroencephalogram (EEG) from awake behaving wildtype and Sp4 hypomorphic mice. We characterized cortical theta-band power and phase-coupling phenotypes, a known neural circuit substrate underlying cognitive functions, and further studied the effects of a subanesthetic dosage of ketamine on theta abnormalities unique to Sp4 hypomorphism. Sp4 hypomorphic mice had markedly elevated theta power localized frontally and parietally, a more pronounced theta phase progression along the neuraxis, and a stronger frontal-parietal theta coupling. Acute subanesthetic ketamine did not affect theta power in wildtype animals but significantly reduced it in Sp4 hypomorphic mice, nearly completely neutralizing their excessive frontal/parietal theta power. Ketamine did not significantly alter cortical theta phase progression in either wildtype or Sp4 hypomorphic animals, but significantly strengthened cortical theta phase-coupling in wildtype, but not in Sp4 hypomorphic animals. Our results suggested that the resting-state phenotypes of cortical theta oscillations unique to Sp4 hypomorphic mice closely mimicked a schizophrenic endophenotype. Further, ketamine independently modulated Sp4 hypomorphic anomalies in theta power and phase-coupling, suggesting separate underlying neural circuit mechanisms.
Collapse
Affiliation(s)
- Xin Wang
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail:
| | - António Pinto-Duarte
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - M. Margarita Behrens
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Xianjin Zhou
- Department of Psychiatry, University of California at San Diego, La Jolla, California, United States of America
| | - Terrence J. Sejnowski
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
- Division of Biology, University of California at San Diego, La Jolla, California, United States of America
| |
Collapse
|
17
|
Ledda F, Paratcha G. Mechanisms regulating dendritic arbor patterning. Cell Mol Life Sci 2017; 74:4511-4537. [PMID: 28735442 PMCID: PMC11107629 DOI: 10.1007/s00018-017-2588-8] [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: 12/04/2016] [Revised: 06/14/2017] [Accepted: 07/06/2017] [Indexed: 12/17/2022]
Abstract
The nervous system is populated by diverse types of neurons, each of which has dendritic trees with strikingly different morphologies. These neuron-specific morphologies determine how dendritic trees integrate thousands of synaptic inputs to generate different firing properties. To ensure proper neuronal function and connectivity, it is necessary that dendrite patterns are precisely controlled and coordinated with synaptic activity. Here, we summarize the molecular and cellular mechanisms that regulate the formation of cell type-specific dendrite patterns during development. We focus on different aspects of vertebrate dendrite patterning that are particularly important in determining the neuronal function; such as the shape, branching, orientation and size of the arbors as well as the development of dendritic spine protrusions that receive excitatory inputs and compartmentalize postsynaptic responses. Additionally, we briefly comment on the implications of aberrant dendritic morphology for nervous system disease.
Collapse
Affiliation(s)
- Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Paraguay 2155, 3rd Floor, CABA, 1121, Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Paraguay 2155, 3rd Floor, CABA, 1121, Buenos Aires, Argentina.
| |
Collapse
|
18
|
MacDowell KS, Pinacho R, Leza JC, Costa J, Ramos B, García-Bueno B. Differential regulation of the TLR4 signalling pathway in post-mortem prefrontal cortex and cerebellum in chronic schizophrenia: Relationship with SP transcription factors. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:481-492. [PMID: 28803924 DOI: 10.1016/j.pnpbp.2017.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/21/2022]
Abstract
Alterations in innate immunity may underlie the pathophysiology of schizophrenia (SZ). Toll-like receptor-4 (TLR4) is a master element of innate immunity. The specificity proteins (SPs), transcription factors recently implicated in SZ, are putative regulatory agents of this. This work was aimed at describing alterations in the TLR4 signalling pathway in postmortem brain prefrontal cortex (PFC) and cerebellum (CB) of 16 chronic SZ patients and 14 controls. The possible association of TLR4 pathway with SP1 and SP4 and SZ negative symptomatology is explored. In PFC, TLR4/myeloid differentiation factor 88 (MyD88)/inhibitory subunit of nuclear factor kappa B alpha (IκBα) protein levels were lower in SZ patients, while nuclear transcription factor-κB (NFκB) activity, cyclooxygenase-2 (COX-2) expression and the lipid peroxidation index malondialdehyde (MDA) appeared increased. The pattern of changes in CB is opposite, except for COX-2 expression that remained augmented and MDA levels unaltered. Network interaction analysis showed that TLR4/MyD88/IκBα/NFκB/COX-2 pathway was coupled in PFC and uncoupled in CB. SP4 co-expressed with TLR4 and NFκB in PFC and both SP1 and SP4 co-expressed with NFκB in CB. In PFC, correlation analysis found an inverse relationship between NFκB and negative symptoms. In summary, we found brain region-specific alterations in the TLR4 signalling pathway in chronic SZ, in which SP transcription factors could participate at different levels. Further studies are required to elucidate the regulatory mechanisms of innate immunity in SZ and its relationship with symptoms.
Collapse
Affiliation(s)
- Karina S MacDowell
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain
| | - Raquel Pinacho
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| | - Juan C Leza
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain
| | - Joan Costa
- Banc de Teixits Neurologics, Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, 08830 Barcelona, Spain; Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
| | - Belén Ramos
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain; Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain; Dept. de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain.
| | - Borja García-Bueno
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain.
| |
Collapse
|
19
|
Majewski Ł, Maciąg F, Boguszewski PM, Wasilewska I, Wiera G, Wójtowicz T, Mozrzymas J, Kuznicki J. Overexpression of STIM1 in neurons in mouse brain improves contextual learning and impairs long-term depression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:1071-1087. [PMID: 27913207 DOI: 10.1016/j.bbamcr.2016.11.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/25/2016] [Accepted: 11/26/2016] [Indexed: 10/20/2022]
Abstract
STIM1 is an endoplasmic reticulum calcium sensor that is involved in several processes in neurons, including store-operated calcium entry. STIM1 also inhibits voltage-gated calcium channels, such as Cav1.2 and Cav3.1, and is thus considered a multifunctional protein. The aim of this work was to investigate the ways in which transgenic neuronal overexpression of STIM1 in FVB/NJ mice affects animal behavior and the electrophysiological properties of neurons in acute hippocampal slices. We overexpressed STIM1 from the Thy1.2 promoter and verified neuronal expression by quantitative reverse-transcription polymerase chain reaction, Western blot, and immunohistochemistry. Mature primary hippocampal cultures expressed STIM1 but exhibited no changes in calcium homeostasis. Basal synaptic transmission efficiency and short-term plasticity were comparable in slices that were isolated from transgenic mice, similarly as the magnitude of long-term potentiation. However, long-term depression that was induced by the glutamate receptor 1/5 agonist (S)-3,5-dihydroxyphenylglycine was impaired in STIM1 slices. Interestingly, transgenic mice exhibited a decrease in anxiety-like behavior and improvements in contextual learning. In summary, our data indicate that STIM1 overexpression in neurons in the brain perturbs metabotropic glutamate receptor signaling, leading to impairments in long-term depression and alterations in animal behavior. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Łukasz Majewski
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., 02-109 Warsaw, Poland
| | - Filip Maciąg
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., 02-109 Warsaw, Poland
| | - Paweł M Boguszewski
- Laboratory of Animal Models, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland
| | - Iga Wasilewska
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., 02-109 Warsaw, Poland
| | - Grzegorz Wiera
- Laboratory of Neuroscience, Dept. Biophysics, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland; Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, 30 Cybulskiego Str., 50-205 Wroclaw, Poland
| | - Tomasz Wójtowicz
- Laboratory of Neuroscience, Dept. Biophysics, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland
| | - Jerzy Mozrzymas
- Laboratory of Neuroscience, Dept. Biophysics, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland; Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, 30 Cybulskiego Str., 50-205 Wroclaw, Poland
| | - Jacek Kuznicki
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., 02-109 Warsaw, Poland.
| |
Collapse
|
20
|
Pinacho R, Villalmanzo N, Meana JJ, Ferrer I, Berengueras A, Haro JM, Villén J, Ramos B. Altered CSNK1E, FABP4 and NEFH protein levels in the dorsolateral prefrontal cortex in schizophrenia. Schizophr Res 2016; 177:88-97. [PMID: 27236410 DOI: 10.1016/j.schres.2016.04.050] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 03/15/2016] [Accepted: 04/27/2016] [Indexed: 11/28/2022]
Abstract
Schizophrenia constitutes a complex disease. Negative and cognitive symptoms are enduring and debilitating components of the disorder, highly associated to disability and burden. Disrupted neurotransmission circuits in dorsolateral prefrontal cortex (DLPFC) have been related to these symptoms. To identify candidates altered in schizophrenia, we performed a pilot proteomic analysis on postmortem human DLPFC tissue from patients with schizophrenia (n=4) and control (n=4) subjects in a pool design using differential isotope peptide labelling followed by liquid chromatography tandem mass spectrometry (LC-MS/MS). We quantified 1315 proteins with two or more unique peptides, 116 of which showed altered changes. Of these altered proteins, we selected four with potential roles on cell signaling, neuronal development and synapse functioning for further validation: casein kinase I isoform epsilon (CSNK1E), fatty acid-binding protein 4 (FABP4), neurofilament triplet H protein (NEFH), and retinal dehydrogenase 1 (ALDH1A1). Immunoblot validation confirmed our proteomic findings of these proteins being decreased in abundance in the schizophrenia samples. Additionally, we conducted immunoblot validation of these candidates on an independent sample cohort comprising 23 patients with chronic schizophrenia and 23 matched controls. In this second cohort, CSNK1E, FABP4 and NEFH were reduced in the schizophrenia group while ALDH1A1 did not significantly change. This study provides evidence indicating these proteins are decreased in schizophrenia: CSNK1E, involved in circadian molecular clock signaling, FABP4 with possible implication in synapse functioning, and NEFH, important for cytoarchitecture organization. Hence, these findings suggest the possible implication of these proteins in the cognitive and/or negative symptoms in schizophrenia.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - Núria Villalmanzo
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - J Javier Meana
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Instituto BioCruces, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Bº Sarriena s/n, 48940 Leioa, Bizkaia, Spain
| | - Isidre Ferrer
- Instituto de Neuropatología, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Centro de Investigación Biomédica en Red para enfermedades neurodegenerativas, CIBERNED, Feixa Llarga s/n, Hospitalet de LLobregat, 08907 Barcelona, Spain
| | - Adriana Berengueras
- Banc de Teixits Neurologics, Parc Sanitari Sant Joan de Déu, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - Josep M Haro
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain
| | - Judit Villén
- Genome Sciences Department, School of Medicine, University of Washington, 3720 15th Ave NE, Seattle 98195, WA, USA
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain.
| |
Collapse
|
21
|
Pinacho R, Vila E, Prades R, Tarragó T, Castro E, Ferrer I, Ramos B. The glial phosphorylase of glycogen isoform is reduced in the dorsolateral prefrontal cortex in chronic schizophrenia. Schizophr Res 2016; 177:37-43. [PMID: 27156240 DOI: 10.1016/j.schres.2016.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Abstract
Reduced glutamatergic activity and energy metabolism in the dorsolateral prefrontal cortex (DLPFC) have been described in schizophrenia. Glycogenolysis in astrocytes is responsible for providing neurons with lactate as a transient energy supply helping to couple glutamatergic neurotransmission and glucose utilization in the brain. This mechanism could be disrupted in schizophrenia. The aim of this study was to explore whether the protein levels of the astrocyte isoform of glycogen phosphorylase (PYGM), key enzyme of glycogenolysis, and the isoform A of Ras-related C3 botulinum toxin substrate 1 (RAC1), a kinase that regulates PYGM activity, are altered in the postmortem DLPFC of chronic schizophrenia patients (n=23) and matched controls (n=23). We also aimed to test NMDAR blockade effect on these proteins in the mouse cortex and cortical astrocytes and antipsychotic treatments in rats. Here we report a reduction in PYGM and RAC1 protein levels in the DLPFC in schizophrenia. We found that treatment with the NMDAR antagonist dizocilpine in mice as a model of psychosis increased PYGM and reduced RAC1 protein levels. The same result was observed in rat cortical astroglial-enriched cultures. 21-day haloperidol treatment increased PYGM levels in rats. These results show that PYGM and RAC1 are altered in the DLPFC in chronic schizophrenia and are controlled by NMDA signalling in the rodent cortex and cortical astrocytes suggesting an altered NMDA-dependent glycogenolysis in astrocytes in schizophrenia. Together, this study provides evidence of a NMDA-dependent transient local energy deficit in neuron-glia crosstalk in schizophrenia, contributing to energy deficits of the disorder.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830, Sant Boi de Llobregat, Barcelona, Spain
| | - Elia Vila
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830, Sant Boi de Llobregat, Barcelona, Spain
| | - Roger Prades
- Iproteos S.L., Baldiri I Reixac, 10, 08028 Barcelona, Spain
| | - Teresa Tarragó
- Iproteos S.L., Baldiri I Reixac, 10, 08028 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), Baldiri I Reixac, 10, 08028 Barcelona, Spain
| | - Elena Castro
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), Departamento de Fisiología y Farmacología, Universidad de Cantabria, 39011, Santander, Spain, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Isidre Ferrer
- Instituto de Neuropatología, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Centro de Investigación Biomédica en Red para enfermedades neurodegenerativas, CIBERNED, Feixa Llarga s/n, 08907 Hospitalet de LLobregat, Barcelona, Spain
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830, Sant Boi de Llobregat, Barcelona, Spain.
| |
Collapse
|
22
|
Papavassiliou KA, Papavassiliou AG. Transcription Factor Drug Targets. J Cell Biochem 2016; 117:2693-2696. [PMID: 27191703 DOI: 10.1002/jcb.25605] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 01/09/2023]
Abstract
Transcription factors represent the point of convergence of multiple signaling pathways within eukaryotic cells. Deregulated transcription factors contribute to the pathogenesis of a plethora of human diseases, ranging from diabetes, inflammatory disorders and cardiovascular disease to many cancers, and thus these proteins hold great therapeutic potential. Direct modulation of transcription factor function by small molecules is no longer regarded a Sisyphean task, as recent work in drug discovery has revealed that transcription factors are amenable to drug inhibition. Here in we summarize, recent advances regarding the significance of transcription factors in human diseases and we discuss emerging pharmacological strategies to modulate transcription factor function. J. Cell. Biochem. 117: 2693-2696, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kostas A Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
23
|
Fusté M, Meléndez-Pérez I, Villalta-Gil V, Pinacho R, Villalmanzo N, Cardoner N, Menchón JM, Haro JM, Soriano-Mas C, Ramos B. Specificity proteins 1 and 4, hippocampal volume and first-episode psychosis. Br J Psychiatry 2016; 208:591-2. [PMID: 26541691 DOI: 10.1192/bjp.bp.114.152140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 11/03/2014] [Indexed: 11/23/2022]
Abstract
We assessed specificity protein 1 (SP1) and 4 (SP4) transcription factor levels in peripheral blood mononuclear cells and conducted a voxel-based morphometry analysis on brain structural magnetic resonance images from 11 patients with first-episode psychosis and 14 healthy controls. We found lower SP1 and SP4 levels in patients, which correlated positively with right hippocampal volume. These results extend previous evidence showing that such transcription factors may constitute a molecular pathway to the development of psychosis.
Collapse
Affiliation(s)
- Montserrat Fusté
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Iria Meléndez-Pérez
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Victoria Villalta-Gil
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Raquel Pinacho
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Núria Villalmanzo
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Narcís Cardoner
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - José M Menchón
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Josep Maria Haro
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Carles Soriano-Mas
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| | - Belén Ramos
- Montserrat Fusté, MBBS, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Iria Meléndez-Pérez, MSc, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Victoria Villalta-Gil, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain and Affective Neuroscience Laboratory, Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA; Raquel Pinacho, MSc, Núria Villalmanzo, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Narcís Cardoner, MD, PhD, José M. Menchón, MD, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Clinical Sciences, School of Medicine, University of Barcelona, Spain; Josep Maria Haro, MD, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain; Carles Soriano-Mas, PhD, Bellvitge Biomedical Research Institute-IDIBELL, Psychiatry Department, Bellvitge University Hospital, CIBERSAM and Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain; Belén Ramos, PhD, Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, CIBERSAM, Sant Boi de Llobregat (Barcelona), Spain
| |
Collapse
|
24
|
Young JW, Kamenski ME, Higa KK, Light GA, Geyer MA, Zhou X. GlyT-1 Inhibition Attenuates Attentional But Not Learning or Motivational Deficits of the Sp4 Hypomorphic Mouse Model Relevant to Psychiatric Disorders. Neuropsychopharmacology 2015; 40:2715-26. [PMID: 25907107 PMCID: PMC4864647 DOI: 10.1038/npp.2015.120] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 01/29/2023]
Abstract
Serious mental illness occurs in 25% of the general population, with many disorders being neurodevelopmental, lifelong, and debilitating. The wide variation and overlap in symptoms across disorders increases the difficulty of research and treatment development. The NIMH Research Domain of Criteria initiative aims to improve our understanding of the molecular and behavioral consequences of specific neurodevelopmental mechanisms across disorders, enabling targeted treatment development. The transcription factor Specificity Protein 4 (SP4) is important for neurodevelopment and is genetically associated with both schizophrenia and bipolar disorder. Reduced Sp4 expression in mice (hypomorphic) reproduces several characteristics of psychiatric disorders. We further tested the utility of Sp4 hypomorphic mice as a model organism relevant to psychiatric disorders by assessing cognitive control plus effort and decision-making aspects of approach motivation using cross-species-relevant tests. Sp4 hypomorphic mice exhibited impaired attention as measured by the 5-Choice Continuous Performance Test, an effect that was attenuated by glycine type-1 transporter (GlyT-1) inhibition. Hypomorphic mice also exhibited reduced motivation to work for a reward and impaired probabilistic learning. These deficits may stem from affected anticipatory reward, analogous to anhedonia in patients with schizophrenia and other psychiatric disorders. Neither positive valence deficit was attenuated by GlyT-1 treatment, suggesting that these and the attentional deficits stem from different underlying mechanisms. Given the association of SP4 gene with schizophrenia and bipolar disorder, the present studies provide support that personalized GlyT-1 inhibition may treat attentional deficits in neuropsychiatric patients with low SP4 levels.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Research Service, VA San Diego Healthcare System, San Diego, CA, USA,Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA, Tel: +1 619 543 3582, Fax: +1 619 735 9205, E-mail:
| | - Mary E Kamenski
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Kerin K Higa
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Gregory A Light
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Mark A Geyer
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Xianjin Zhou
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
25
|
Pinacho R, Saia G, Meana JJ, Gill G, Ramos B. Transcription factor SP4 phosphorylation is altered in the postmortem cerebellum of bipolar disorder and schizophrenia subjects. Eur Neuropsychopharmacol 2015; 25:1650-1660. [PMID: 26049820 PMCID: PMC4600646 DOI: 10.1016/j.euroneuro.2015.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 12/12/2014] [Accepted: 05/13/2015] [Indexed: 11/27/2022]
Abstract
Transcription factors play important roles in the control of neuronal function in physiological and pathological conditions. We previously reported reduced levels of transcription factor SP4 protein, but not transcript, in the cerebellum in bipolar disorder and associated with more severe negative symptoms in schizophrenia. We have recently reported phosphorylation of Sp4 at S770, which is regulated by membrane depolarization and NMDA receptor activity. The aim of this study was to investigate SP4 S770 phosphorylation in bipolar disorder and its association with negative symptoms in schizophrenia, and to explore the potential relationship between phosphorylation and protein abundance. Here we report a significant increase in SP4 phosphorylation in the cerebellum, but not the prefrontal cortex, of bipolar disorder subjects (n=10) (80% suicide) compared to matched controls (n=10). We found that SP4 phosphorylation inversely correlated with SP4 levels independently of disease status in both areas of the human brain. Moreover, SP4 phosphorylation in the cerebellum positively correlated with negative symptoms in schizophrenia subjects (n=15). Further, we observed that a phospho-mimetic mutation in truncated Sp4 was sufficient to significantly decrease Sp4 steady-state levels, while a non-phosphorylatable mutant showed increased stability in cultured rat cerebellar granule neurons. Our results indicate that SP4 S770 phosphorylation is increased in the cerebellum in bipolar disorder subjects that committed suicide and in severe schizophrenia subjects, and may be part of a degradation signal that controls Sp4 abundance in cerebellar granule neurons. This opens the possibility that modulation of SP4 phosphorylation may contribute to the molecular pathophysiology of psychotic disorders.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830- Sant Boi de Llobregat, Barcelona, Spain
| | - Gregory Saia
- Department of Developmental, Molecular, and Chemical Biology, Sackler School of Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111.,Cell, Molecular and Developmental Biology Program, Sackler School of Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111
| | - J Javier Meana
- Departamento de Farmacología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, B Sarriena s/n 48940-Leioa, Bizkaia, Spain
| | - Grace Gill
- Department of Developmental, Molecular, and Chemical Biology, Sackler School of Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830- Sant Boi de Llobregat, Barcelona, Spain
| |
Collapse
|
26
|
Majewski L, Kuznicki J. SOCE in neurons: Signaling or just refilling? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1940-52. [DOI: 10.1016/j.bbamcr.2015.01.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 01/14/2023]
|
27
|
Sp3/REST/HDAC1/HDAC2 Complex Represses and Sp1/HIF-1/p300 Complex Activates ncx1 Gene Transcription, in Brain Ischemia and in Ischemic Brain Preconditioning, by Epigenetic Mechanism. J Neurosci 2015; 35:7332-48. [PMID: 25972164 DOI: 10.1523/jneurosci.2174-14.2015] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Na(+)-Ca(2+) exchanger 1 (NCX1) is reduced in stroke by the RE1-silencing transcription factor (REST), whereas it is increased in ischemic brain preconditioning (PC) by hypoxia-inducible factor 1 (HIF-1). Because ncx1 brain promoter (ncx1-Br) has five putative consensus sequences, named Sp1A-E, for the specificity protein (Sp) family of transcription factors (Sp1-4), we investigated the role of this family in regulating ncx1 transcription in rat cortical neurons. Here we found that Sp1 is a transcriptional activator, whereas Sp3 is a transcriptional repressor of ncx1, and that both bind ncx1-Br in a sequence-specific manner, modulating ncx1 transcription through the Sp1 sites C-E. Furthermore, by transient middle cerebral artery occlusion (tMCAO) in rats, the transcriptional repressors Sp3 and REST colocalized with the two histone-deacetylases (HDACs) HDAC1 and HDAC2 on the ncx1-Br, with a consequent hypoacetylation. Contrarily, in PC+tMCAO the transcriptional activators Sp1 and HIF-1 colocalized with histone acetyltransferase p300 on ncx1-Br with a consequent hyperacetylation. In addition, in neurons silenced with siRNA of NCX1 and subjected to oxygen and glucose deprivation (OGD) (3 h) plus reoxygenation (RX) (24 h), the neuroprotection of Class I HDAC inhibitor MS-275 was counteracted, whereas in neurons overexpressing NCX1 and subjected to ischemic preconditioning (PC+OGD/RX), the neurotoxic effect of p300 inhibitor C646 was prevented. Collectively, these results demonstrate that NCX1 expression is regulated by the Sp3/REST/HDAC1/HDAC2 complex in tMCAO and by the Sp1/HIF-1/p300 complex in PC+tMCAO and that epigenetic intervention, by modulating the acetylation of ncx1-Br, may be a strategy for the development of innovative therapeutic intervention in stroke.
Collapse
|
28
|
Higa KK, Ji B, Buell MR, Risbrough VB, Powell SB, Young JW, Geyer MA, Zhou X. Restoration of Sp4 in Forebrain GABAergic Neurons Rescues Hypersensitivity to Ketamine in Sp4 Hypomorphic Mice. Int J Neuropsychopharmacol 2015; 18:pyv063. [PMID: 26037489 PMCID: PMC4756721 DOI: 10.1093/ijnp/pyv063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/29/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ketamine produces schizophrenia-like behavioral phenotypes in healthy people. Prolonged ketamine effects and exacerbation of symptoms after the administration of ketamine have been observed in patients with schizophrenia. More recently, ketamine has been used as a potent antidepressant to treat patients with major depression. The genes and neurons that regulate behavioral responses to ketamine, however, remain poorly understood. Sp4 is a transcription factor for which gene expression is restricted to neuronal cells in the brain. Our previous studies demonstrated that Sp4 hypomorphic mice display several behavioral phenotypes relevant to psychiatric disorders, consistent with human SP4 gene associations with schizophrenia, bipolar disorder, and major depression. Among those behavioral phenotypes, hypersensitivity to ketamine-induced hyperlocomotion has been observed in Sp4 hypomorphic mice. METHODS In the present study, we used the Cre-LoxP system to restore Sp4 gene expression, specifically in either forebrain excitatory or GABAergic inhibitory neurons in Sp4 hypomorphic mice. Mouse behavioral phenotypes related to psychiatric disorders were examined in these distinct rescue mice. RESULTS Restoration of Sp4 in forebrain excitatory neurons did not rescue deficient sensorimotor gating nor ketamine-induced hyperlocomotion. Restoration of Sp4 in forebrain GABAergic neurons, however, rescued ketamine-induced hyperlocomotion, but did not rescue deficient sensorimotor gating. CONCLUSIONS Our studies suggest that the Sp4 gene in forebrain GABAergic neurons regulates ketamine-induced hyperlocomotion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xianjin Zhou
- Department of Psychiatry, University of California San Diego, La Jolla, CA (Ms Higa, Drs Ji, Risbrough, Powell, Young, Geyer, and Zhou, and Ms Buell); Research Service, VA San Diego Healthcare System, La Jolla, CA (Drs Risbrough, Powell, Young, Geyer, and Zhou, and Ms Buell); Neurosciences Graduate Program, University of California San Diego, La Jolla, CA (Ms Higa).
| |
Collapse
|
29
|
Pinacho R, Saia G, Fusté M, Meléndez-Pérez I, Villalta-Gil V, Haro JM, Gill G, Ramos B. Phosphorylation of transcription factor specificity protein 4 is increased in peripheral blood mononuclear cells of first-episode psychosis. PLoS One 2015; 10:e0125115. [PMID: 25915526 PMCID: PMC4411105 DOI: 10.1371/journal.pone.0125115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/20/2015] [Indexed: 12/20/2022] Open
Abstract
Background Altered expression of transcription factor specificity protein 4 (SP4) has been found in the postmortem brain of patients with psychiatric disorders including schizophrenia and bipolar disorder. Reduced levels of SP4 protein have recently been reported in peripheral blood mononuclear cells in first-episode psychosis. Also, SP4 levels are modulated by lithium treatment in cultured neurons. Phosphorylation of SP4 at S770 is increased in the cerebellum of bipolar disorder subjects and upon inhibition of NMDA receptor signaling in cultured neurons. The aim of this study was to investigate whether SP4 S770 phosphorylation is increased in lymphocytes of first-episode psychosis patients and the effect of lithium treatment on this phosphorylation. Methods A cross-sectional study of S770 phosphorylation relative to total SP4 immunoreactivity using specific antibodies in peripheral blood mononuclear cells in first-episode psychosis patients (n = 14, treated with lithium or not) and matched healthy controls (n = 14) by immunoblot was designed. We also determined the effects of the prescribed drugs lithium, olanzapine or valproic acid on SP4 phosphorylation in rat primary cultured cerebellar granule neurons. Results We found that SP4 S770 phosphorylation was significantly increased in lymphocytes in first-episode psychosis compared to controls and decreased in patients treated with lithium compared to patients who did not receive lithium. Moreover, incubation with lithium but not olanzapine or valproic acid reduced SP4 phosphorylation in rat cultured cerebellar granule neurons. Conclusions The findings presented here indicate that SP4 S770 phosphorylation is increased in lymphocytes in first-episode psychosis which may be reduced by lithium treatment in patients. Moreover, our study shows lithium treatment prevents this phosphorylation in vitro in neurons. This pilot study suggests that S770 SP4 phosphorylation could be a peripheral biomarker of psychosis, and may be regulated by lithium treatment in first-episode psychosis.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Fundació Sant Joan de Déu, Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
| | - Gregory Saia
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Cell, Molecular and Developmental Biology Program, Sackler School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Montserrat Fusté
- Unitat de recerca, Fundació Sant Joan de Déu, Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
| | - Iria Meléndez-Pérez
- Unitat de recerca, Fundació Sant Joan de Déu, Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
| | - Victoria Villalta-Gil
- Unitat de recerca, Fundació Sant Joan de Déu, Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
| | - Josep Maria Haro
- Unitat de recerca, Fundació Sant Joan de Déu, Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
| | - Grace Gill
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (BR); (GG)
| | - Belén Ramos
- Unitat de recerca, Fundació Sant Joan de Déu, Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
- * E-mail: (BR); (GG)
| |
Collapse
|
30
|
Moccia F, Zuccolo E, Soda T, Tanzi F, Guerra G, Mapelli L, Lodola F, D'Angelo E. Stim and Orai proteins in neuronal Ca(2+) signaling and excitability. Front Cell Neurosci 2015; 9:153. [PMID: 25964739 PMCID: PMC4408853 DOI: 10.3389/fncel.2015.00153] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/03/2015] [Indexed: 02/01/2023] Open
Abstract
Stim1 and Orai1 are ubiquitous proteins that have long been known to mediate Ca2+ release-activated Ca2+ (CRAC) current (ICRAC) and store-operated Ca2+ entry (SOCE) only in non-excitable cells. SOCE is activated following the depletion of the endogenous Ca2+ stores, which are mainly located within the endoplasmic reticulum (ER), to replete the intracellular Ca2+ reservoir and engage specific Ca2+-dependent processes, such as proliferation, migration, cytoskeletal remodeling, and gene expression. Their paralogs, Stim2, Orai2 and Orai3, support SOCE in heterologous expression systems, but their physiological role is still obscure. Ca2+ inflow in neurons has long been exclusively ascribed to voltage-operated and receptor-operated channels. Nevertheless, recent work has unveiled that Stim1–2 and Orai1-2, but not Orai3, proteins are also expressed and mediate SOCE in neurons. Herein, we survey current knowledge about the neuronal distribution of Stim and Orai proteins in rodent and human brains; we further discuss that Orai2 is the main pore-forming subunit of CRAC channels in central neurons, in which it may be activated by either Stim1 or Stim2 depending on species, brain region and physiological stimuli. We examine the functions regulated by SOCE in neurons, where this pathway is activated under resting conditions to refill the ER, control spinogenesis and regulate gene transcription. Besides, we highlighted the possibility that SOCE also controls neuronal excitation and regulate synaptic plasticity. Finally, we evaluate the involvement of Stim and Orai proteins in severe neurodegenerative and neurological disorders, such as Alzheimer’s disease and epilepsy.
Collapse
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Teresa Soda
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy
| | - Franco Tanzi
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Lisa Mapelli
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Roma, Italy
| | - Francesco Lodola
- Laboratory of Molecular Cardiology, IRCCS Fondazione Salvatore Maugeri Pavia, Italy
| | - Egidio D'Angelo
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Brain Connectivity Center, C. Mondino National Neurological Institute, Fondazione IRCCS Policlinico San Matteo Pavia Pavia, Italy
| |
Collapse
|
31
|
Pinacho R, Valdizán EM, Pilar-Cuellar F, Prades R, Tarragó T, Haro JM, Ferrer I, Ramos B. Increased SP4 and SP1 transcription factor expression in the postmortem hippocampus of chronic schizophrenia. J Psychiatr Res 2014; 58:189-96. [PMID: 25175639 DOI: 10.1016/j.jpsychires.2014.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/08/2014] [Accepted: 08/08/2014] [Indexed: 12/20/2022]
Abstract
Altered levels of transcription factor specificity protein 4 (SP4) and 1 (SP1) in the cerebellum, prefrontal cortex and/or lymphocytes have been reported in severe psychiatric disorders, including early psychosis, bipolar disorder, and chronic schizophrenia subjects who have undergone long-term antipsychotic treatments. SP4 transgenic mice show altered hippocampal-dependent psychotic-like behaviours and altered development of hippocampal dentate gyrus. Moreover, NMDAR activity regulates SP4 function. The aim of this study was to investigate SP4 and SP1 expression levels in the hippocampus in schizophrenia, and the possible effect of antipsychotics and NMDAR blockade on SP protein levels in rodent hippocampus. We analysed SP4 and SP1 expression levels in the postmortem hippocampus of chronic schizophrenia (n = 14) and control (n = 11) subjects by immunoblot and quantitative RT-PCR. We tested the effect of NMDAR blockade on SP factors in the hippocampus of mouse treated with an acute dose of MK801. We also investigated the effect of subacute treatments with haloperidol and clozapine on SP protein levels in the rat hippocampus. We report that SP4 protein and both SP4 and SP1 mRNA expression levels are significantly increased in the hippocampus in chronic schizophrenia. Likewise, acute treatment with MK801 increased both SP4 and SP1 protein levels in mouse hippocampus. In contrast, subacute treatment with haloperidol and clozapine did not significantly alter SP protein levels in rat hippocampus. These results suggest that SP4 and SP1 upregulation may be part of the mechanisms deregulated downstream of glutamate signalling pathways in schizophrenia and might be contributing to the hippocampal-dependent cognitive deficits of the disorder.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain
| | - Elsa M Valdizán
- Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC-SODERCAN), Departamento de Fisiología y Farmacología, Universidad de Cantabria, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Avda. Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Fuencisla Pilar-Cuellar
- Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC-SODERCAN), Departamento de Fisiología y Farmacología, Universidad de Cantabria, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Avda. Cardenal Herrera Oria s/n, 39011, Santander, Spain
| | - Roger Prades
- Iproteos S.L., Baldiri I Reixac, 10, 08028 Barcelona, Spain
| | - Teresa Tarragó
- Iproteos S.L., Baldiri I Reixac, 10, 08028 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), Baldiri I Reixac, 10, 08028 Barcelona, Spain
| | - Josep Maria Haro
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain
| | - Isidre Ferrer
- Instituto de Neuropatología, IDIBELL-Hospital Universitario de Bellvitge, Universitat de Barcelona, Centro de Investigación Biomédica en Red para enfermedades neurodegenerativas, CIBERNED. Feixa Llarga s/n, 08907 Hospitalet de LLobregat, Barcelona, Spain
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain.
| |
Collapse
|
32
|
Genetics of psychotropic medication induced side effects in two independent samples of bipolar patients. J Neural Transm (Vienna) 2014; 122:43-58. [PMID: 25129258 DOI: 10.1007/s00702-014-1290-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/02/2014] [Indexed: 02/07/2023]
Abstract
The treatment of bipolar disorder (BD) usually requires combination therapies, with the critical issue of the emergence of adverse drug reactions (ADRs) and the possibility of low treatment adherence. Genetic polymorphisms are hypothesized to modulate the pharmacodynamics of psychotropic drugs, representing potential biological markers of ADRs. This study investigated genes involved in the regulation of neuroplasticity (BDNF, ST8SIA2), second messenger cascades (GSK3B, MAPK1, and CREB1), circadian rhythms (RORA), transcription (SP4, ZNF804A), and monoaminergic system (HTR2A and COMT) in the risk of neurological, psychic, autonomic, and other ADRs. Two independent samples of BD patients naturalistically treated were included (COPE-BD n = 147; STEP-BD n = 659). In the COPE-BD 34 SNPs were genotyped, while in the STEP-BD polymorphisms in the selected genes were extracted from the genome-wide dataset. Each ADRs group was categorized as absent-mild or moderate-severe and logistic regression with appropriate covariates was applied to identify possible risk genotypes/alleles. 58.5 and 93.5 % of patients were treated with mood stabilizers, 44.2 and 50.7 % were treated with antipsychotics, and 69.4 and 46.1 % were treated with antidepressants in the COPE-BD and STEP-BD, respectively. Our findings suggested that ST8SIA2 may be associated with psychic ADRs, as shown in the COPE-BD (rs4777989 p = 0.0017) and STEP-BD (rs56027313, rs13379489 and rs10852173). A cluster of RORA SNPs around rs2083074 showed an effect on psychic ADRs in the STEP-BD. Trends supporting the association between HTR2A and autonomic ADRs were found in both samples. Confirmations are needed particularly for ST8SIA2 and RORA since the few available data regarding their role in relation to psychotropic ADRs.
Collapse
|
33
|
Sun X, Pinacho R, Saia G, Punko D, Meana JJ, Ramos B, Gill G. Transcription factor Sp4 regulates expression of nervous wreck 2 to control NMDAR1 levels and dendrite patterning. Dev Neurobiol 2014; 75:93-108. [PMID: 25045015 DOI: 10.1002/dneu.22212] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 06/20/2014] [Accepted: 07/13/2014] [Indexed: 02/06/2023]
Abstract
Glutamatergic signaling through N-methyl-d-aspartate receptors (NMDARs) is important for neuronal development and plasticity and is often dysregulated in psychiatric disorders. Mice mutant for the transcription factor Sp4 have reduced levels of NMDAR subunit 1 (NR1) protein, but not mRNA, and exhibit behavioral and memory deficits (Zhou et al., [2010] Human Molecular Genetics 19: 3797-3805). In developing cerebellar granule neurons (CGNs), Sp4 controls dendrite patterning (Ramos et al., [2007] Proc Natl Acad Sci USA 104: 9882-9887). Sp4 target genes that regulate dendrite pruning or NR1 levels are not known. Here we report that Sp4 activates transcription of Nervous Wreck 2 (Nwk2; also known as Fchsd1) and, further, that Nwk2, an F-BAR domain-containing protein, mediates Sp4-dependent regulation of dendrite patterning and cell surface expression of NR1. Knockdown of Nwk2 in CGNs increased primary dendrite number, phenocopying Sp4 knockdown, and exogenous expression of Nwk2 in Sp4-depleted neurons rescued dendrite number. We observed that acute Sp4 depletion reduced levels of surface, but not total, NR1, and this was rescued by Nwk2 expression. Furthermore, expression of Nr1 suppressed the increase in dendrite number in Sp4- or Nwk2- depleted neurons. We previously reported that Sp4 protein levels were reduced in cerebellum of subjects with bipolar disorder (BD) (Pinacho et al., [2011] Bipolar Disorders 13: 474-485). Here we report that Nwk2 mRNA and NR1 protein levels were also reduced in postmortem cerebellum of BD subjects. Our data suggest a role for Sp4-regulated Nwk2 in NMDAR trafficking and identify a Sp4-Nwk2-NMDAR1 pathway that regulates neuronal morphogenesis during development and may be disrupted in bipolar disorder.
Collapse
Affiliation(s)
- Xinxin Sun
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, 02111; Genetics Program, Sackler School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, 02111
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Almost a decade has passed since first STIM, and later Orai, proteins were identified as the molecular constituents of store-operated calcium entry (SOCE). Their roles in immune function have been intensely investigated, but the roles of STIM and Orai in neuronal cells have been much less clear. Lalonde et al. show that when neurons are hyperpolarized or "at rest," constitutive endoplasmic reticulum (ER) Ca(2+) release leads to SOCE-mediated activation of neuronal transcription factors. Precisely why ER Ca(2+) release is constitutive in neurons remains an important question. Irrespective of the answer, this observation provides an intriguing new perspective on why a relatively low-abundance, small-conductance channel such as Orai1 would be important in neurons, which contain a relative abundance of voltage-operated Ca(2+) channels.
Collapse
Affiliation(s)
- Robert Hooper
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Brad S Rothberg
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jonathan Soboloff
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
35
|
Lalonde J, Saia G, Gill G. Store-operated calcium entry promotes the degradation of the transcription factor Sp4 in resting neurons. Sci Signal 2014; 7:ra51. [PMID: 24894994 DOI: 10.1126/scisignal.2005242] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Calcium (Ca(2+)) signaling activated in response to membrane depolarization regulates neuronal maturation, connectivity, and plasticity. Store-operated Ca(2+) entry (SOCE) occurs in response to depletion of Ca(2+) from endoplasmic reticulum (ER), mediates refilling of this Ca(2+) store, and supports Ca(2+) signaling in nonexcitable cells. We report that maximal activation of SOCE occurred in cerebellar granule neurons cultured under resting conditions and that this Ca(2+) influx promoted the degradation of transcription factor Sp4, a regulator of neuronal morphogenesis and function. Lowering the concentration of extracellular potassium, a condition that reduces neuronal excitability, stimulated depletion of intracellular Ca(2+) stores, resulted in the relocalization of the ER Ca(2+) sensor STIM1 into punctate clusters consistent with multimerization and accumulation at junctions between the ER and plasma membrane, and induced a Ca(2+) influx with characteristics of SOCE. Compounds that block SOCE prevented the ubiquitylation and degradation of Sp4 in neurons exposed to a low concentration of extracellular potassium. Knockdown of STIM1 blocked degradation of Sp4, whereas expression of constitutively active STIM1 decreased Sp4 abundance under depolarizing conditions. Our findings indicated that, in neurons, SOCE is induced by hyperpolarization, and suggested that this Ca(2+) influx pathway is a distinct mechanism for regulating neuronal gene expression.
Collapse
Affiliation(s)
- Jasmin Lalonde
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| | - Gregory Saia
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA. Cell, Molecular & Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Grace Gill
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
36
|
Brain Na(+), K(+)-ATPase Activity In Aging and Disease. INTERNATIONAL JOURNAL OF BIOMEDICAL SCIENCE : IJBS 2014; 10:85-102. [PMID: 25018677 PMCID: PMC4092085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/05/2014] [Indexed: 11/15/2022]
Abstract
Na(+)/K(+) pump or sodium- and potassium-activated adenosine 5'-triphosphatase (Na(+), K(+)-ATPase), its enzymatic version, is a crucial protein responsible for the electrochemical gradient across the cell membranes. It is an ion transporter, which in addition to exchange cations, is the ligand for cardenolides. This enzyme regulates the entry of K(+) with the exit of Na(+) from cells, being the responsible for Na(+)/K(+) equilibrium maintenance through neuronal membranes. This transport system couples the hydrolysis of one molecule of ATP to exchange three sodium ions for two potassium ions, thus maintaining the normal gradient of these cations in animal cells. Oxidative metabolism is very active in brain, where large amounts of chemical energy as ATP molecules are consumed, mostly required for the maintenance of the ionic gradients that underlie resting and action potentials which are involved in nerve impulse propagation, neurotransmitter release and cation homeostasis. Protein phosphorylation is a key process in biological regulation. At nervous system level, protein phosphorylation is the major molecular mechanism through which the function of neural proteins is modulted in response to extracellular signals, including the response to neurotransmitter stimuli. It is the major mechanism of neural plasticity, including memory processing. The phosphorylation of Na(+), K(+)-ATPase catalytic subunit inhibits enzyme activity whereas the inhibition of protein kinase C restores the enzyme activity. The dephosphorylation of neuronal Na(+), K(+)-ATPase is mediated by calcineurin, a serine / threonine phosphatase. The latter enzyme is involved in a wide range of cellular responses to Ca(2+) mobilizing signals, in the regulation of neuronal excitability by controlling the activity of ion channels, in the release of neurotransmitters and hormones, as well as in synaptic plasticity and gene transcription. In the present article evidence showing Na(+), K(+)-ATPase involvement in signaling pathways, enzyme changes in diverse neurological diseases as well as during aging, have been summarized. Issues refer mainly to Na(+), K(+)-ATPase studies in ischemia, brain injury, depression and mood disorders, mania, stress, Alzheimer´s disease, learning and memory, and neuronal hyperexcitability and epilepsy.
Collapse
|
37
|
Saia G, Lalonde J, Sun X, Ramos B, Gill G. Phosphorylation of the transcription factor Sp4 is reduced by NMDA receptor signaling. J Neurochem 2014; 129:743-52. [PMID: 24475768 DOI: 10.1111/jnc.12657] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/06/2014] [Accepted: 01/09/2014] [Indexed: 01/10/2023]
Abstract
The regulation of transcription factor function in response to neuronal activity is important for development and function of the nervous system. The transcription factor Sp4 regulates the developmental patterning of dendrites, contributes to complex processes including learning and memory, and has been linked to psychiatric disorders such as schizophrenia and bipolar disorder. Despite its many roles in the nervous system, the molecular mechanisms regulating Sp4 activity are poorly understood. Here, we report a site of phosphorylation on Sp4 at serine 770 that is decreased in response to membrane depolarization. Inhibition of the voltage-dependent NMDA receptor increased Sp4 phosphorylation. Conversely, stimulation with NMDA reduced the levels of Sp4 phosphorylation, and this was dependent on the protein phosphatase 1/2A. A phosphomimetic substitution at S770 impaired the Sp4-dependent maturation of cerebellar granule neuron primary dendrites, whereas a non-phosphorylatable Sp4 mutant behaved like wild type. These data reveal that transcription factor Sp4 is regulated by NMDA receptor-dependent activation of a protein phosphatase 1/2A signaling pathway. Our findings also suggest that the regulated control of Sp4 activity is an important mechanism governing the developmental patterning of dendrites.
Collapse
Affiliation(s)
- Gregory Saia
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA; the Cell, Molecular and Developmental Biology Program, Sackler School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
38
|
Johar K, Priya A, Wong-Riley MTT. Regulation of Na(+)/K(+)-ATPase by neuron-specific transcription factor Sp4: implication in the tight coupling of energy production, neuronal activity and energy consumption in neurons. Eur J Neurosci 2013; 39:566-78. [PMID: 24219545 DOI: 10.1111/ejn.12415] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 10/03/2013] [Accepted: 10/07/2013] [Indexed: 12/24/2022]
Abstract
A major source of energy demand in neurons is the Na(+)/K(+)-ATPase pump that restores the ionic gradient across the plasma membrane subsequent to depolarizing neuronal activity. The energy comes primarily from mitochondrial oxidative metabolism, of which cytochrome c oxidase (COX) is a key enzyme. Recently, we found that all 13 subunits of COX are regulated by specificity (Sp) factors, and that the neuron-specific Sp4, but not Sp1 or Sp3, regulates the expression of key glutamatergic receptor subunits as well. The present study sought to test our hypothesis that Sp4 also regulates Na(+)/K(+)-ATPase subunit genes in neurons. By means of multiple approaches, including in silico analysis, electrophoretic mobility shift and supershift assays, chromatin immunoprecipitation, promoter mutational analysis, over-expression, and RNA interference studies, we found that Sp4, with minor contributions from Sp1 and Sp3, functionally regulate the Atp1a1, Atp1a3, and Atp1b1 subunit genes of Na(+)/K(+)-ATPase in neurons. Transcripts of all three genes were up-regulated by depolarizing KCl stimulation and down-regulated by the impulse blocker tetrodotoxin (TTX), indicating that their expression was activity-dependent. Silencing of Sp4 blocked the up-regulation of these genes induced by KCl, whereas over-expression of Sp4 rescued them from TTX-induced suppression. The effect of silencing or over-expressing Sp4 on primary neurons was much greater than those of Sp1 or Sp3. The binding sites of Sp factors on these genes are conserved among mice, rats and humans. Thus, Sp4 plays an important role in the transcriptional coupling of energy generation and energy consumption in neurons.
Collapse
Affiliation(s)
- Kaid Johar
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | | | | |
Collapse
|
39
|
Fusté M, Pinacho R, Meléndez-Pérez I, Villalmanzo N, Villalta-Gil V, Haro JM, Ramos B. Reduced expression of SP1 and SP4 transcription factors in peripheral blood mononuclear cells in first-episode psychosis. J Psychiatr Res 2013; 47:1608-14. [PMID: 23941741 DOI: 10.1016/j.jpsychires.2013.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 07/12/2013] [Accepted: 07/16/2013] [Indexed: 12/22/2022]
Abstract
Alterations of transcription factor specificity protein 4 (SP4) and 1 (SP1) have been linked to different neuropsychiatric diseases. Reduced SP4 and SP1 protein levels in the prefrontal cortex have been associated with bipolar disorder and schizophrenia, respectively, suggesting that both factors could be involved in the pathogenesis of disorders with psychotic features. The aim of this study was to investigate whether the reduction of SP1, SP4 and SP3 protein and mRNA expression in peripheral blood mononuclear cells in the early stages of psychosis may act as a potential biomarker of these disorders. A cross-sectional study of first-episode psychosis patients (n = 14) compared to gender- and age-matched healthy controls (n = 14) was designed. Patients were recruited through the adult mental health services of Parc Sanitari Sant Joan de Déu. Protein and gene expression levels of SP1, SP4 and SP3 were assessed in peripheral blood mononuclear cells of patients with first-episode psychosis and healthy control subjects. We report that protein levels of SP1 and SP4, but not SP3, are significantly reduced in patients compared to controls. In contrast, we did not observe any differences in expression levels for SP1, SP4 or SP3 genes between patient and control groups. In patients, SP4 protein levels were significantly associated with SP1 protein levels. No association was found, however, between protein and gene expression levels for each factor. Our study shows reduced SP1 and SP4 protein levels in first-episode psychosis in lymphocytes, suggesting that these transcription factors are potential peripheral biomarkers of psychotic spectrum disorders in the early stages.
Collapse
Affiliation(s)
- Montserrat Fusté
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Pinacho R, Villalmanzo N, Roca M, Iniesta R, Monje A, Haro JM, Meana JJ, Ferrer I, Gill G, Ramos B. Analysis of Sp transcription factors in the postmortem brain of chronic schizophrenia: a pilot study of relationship to negative symptoms. J Psychiatr Res 2013; 47:926-34. [PMID: 23540600 DOI: 10.1016/j.jpsychires.2013.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 03/07/2013] [Accepted: 03/08/2013] [Indexed: 01/13/2023]
Abstract
Negative symptoms are the most resilient manifestations in schizophrenia. An imbalance in dopamine and glutamate pathways has been proposed for the emergence of these symptoms. SP1, SP3 and SP4 transcription factors regulate genes in these pathways, suggesting a possible involvement in negative symptoms. In this study, we characterized Sp factors in the brains of subjects with schizophrenia and explored a possible association with negative symptoms. We also included analysis of NR1, NR2A and DRD2 as Sp target genes. Postmortem cerebellum and prefrontal cortex from an antemortem clinically well-characterized and controlled collection of elderly subjects with chronic schizophrenia (n = 16) and control individuals (n = 14) were examined. We used the Positive and Negative Syndrome and the Clinical Global Impression Schizophrenia scales, quantitative PCR and immunoblot. SP1 protein and mRNA were reduced in the prefrontal cortex in schizophrenia whereas none of Sp factors were altered in the cerebellum. However, we found that SP1, SP3 and SP4 protein levels inversely correlated with negative symptoms in the cerebellum. Furthermore, NR2A and DRD2 mRNA levels correlated with negative symptoms in the cerebellum. In the prefrontal cortex, SP1 mRNA and NR1 and DRD2 inversely correlated with these symptoms while Sp protein levels did not. This pilot study not only reinforces the involvement of SP1 in schizophrenia, but also suggests that reduced levels or function of SP1, SP4 and SP3 may participate in negative symptoms, in part through the regulation of NMDA receptor subunits and/or Dopamine D2 receptor, providing novel information about the complex negative symptoms in this disorder.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de Recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Prolonged Ketamine Effects in Sp4 Hypomorphic Mice: Mimicking Phenotypes of Schizophrenia. PLoS One 2013; 8:e66327. [PMID: 23823008 PMCID: PMC3688895 DOI: 10.1371/journal.pone.0066327] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/04/2013] [Indexed: 12/11/2022] Open
Abstract
It has been well established that schizophrenia patients display impaired NMDA receptor (NMDAR) functions as well as exacerbation of symptoms in response to NMDAR antagonists. Abnormal NMDAR signaling presumably contributes to cognitive deficits which substantially contribute to functional disability in schizophrenia. Establishing a mouse genetic model will help investigate molecular mechanisms of hypoglutmatergic neurotransmission in schizophrenia. Here, we examined the responses of Sp4 hypomorphic mice to NMDAR antagonists in electroencephalography and various behavioral paradigms. Sp4 hypomorphic mice, previously reported to have reduced NMDAR1 expression and LTP deficit in hippocampal CA1, displayed increased sensitivity and prolonged responses to NMDAR antagonists. Molecular studies demonstrated reduced expression of glutamic acid decarboxylase 67 (GAD67) in both cortex and hippocampus, consistent with abnormal gamma oscillations in Sp4 hypomorphic mice. On the other hand, human SP4 gene was reported to be deleted in schizophrenia. Several human genetic studies suggested the association of SP4 gene with schizophrenia and other psychiatric disorders. Therefore, elucidation of the Sp4 molecular pathway in Sp4 hypomorphic mice may provide novel insights to our understanding of abnormal NMDAR signaling in schizophrenia.
Collapse
|