1
|
Vojdani A, Almulla AF, Zhou B, Al-Hakeim HK, Maes M. Reactivation of herpesvirus type 6 and IgA/IgM-mediated responses to activin-A underpin long COVID, including affective symptoms and chronic fatigue syndrome. Acta Neuropsychiatr 2024; 36:172-184. [PMID: 38571295 DOI: 10.1017/neu.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
BACKGROUND Persistent infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), reactivation of dormant viruses, and immune-oxidative responses are involved in long COVID. OBJECTIVES To investigate whether long COVID and depressive, anxiety, and chronic fatigue syndrome (CFS) symptoms are associated with IgA/IgM/IgG to SARS-CoV-2, human herpesvirus type 6 (HHV-6), Epstein-Barr Virus (EBV), and immune-oxidative biomarkers. METHODS We examined 90 long COVID patients and ninety healthy controls. We measured serum IgA/IgM/IgG against HHV-6 and EBV and their deoxyuridine 5′-triphosphate nucleotidohydrolase (duTPase), SARS-CoV-2, and activin-A, C-reactive protein (CRP), advanced oxidation protein products (AOPP), and insulin resistance (HOMA2-IR). RESULTS Long COVID patients showed significant elevations in IgG/IgM-SARS-CoV-2, IgG/IgM-HHV-6, and HHV-6-duTPase, IgA/IgM-activin-A, CRP, AOPP, and HOMA2-IR. Neural network analysis yielded a highly significant predictive accuracy of 80.6% for the long COVID diagnosis (sensitivity: 78.9%, specificity: 81.8%, area under the ROC curve = 0.876); the topmost predictors were as follows: IGA-activin-A, IgG-HHV-6, IgM-HHV-6-duTPase, IgG-SARS-CoV-2, and IgM-HHV-6 (all positively) and a factor extracted from all IgA levels to all viral antigens (inversely). The top 5 predictors of affective symptoms due to long COVID were IgM-HHV-6-duTPase, IgG-HHV-6, CRP, education, IgA-activin-A (predictive accuracy of r = 0.636). The top 5 predictors of CFS due to long COVID were in descending order: CRP, IgG-HHV-6-duTPase, IgM-activin-A, IgM-SARS-CoV-2, and IgA-activin-A (predictive accuracy: r = 0.709). CONCLUSION Reactivation of HHV-6, SARS-CoV-2 persistence, and autoimmune reactions to activin-A combined with activated immune-oxidative pathways play a major role in the pathophysiology of long COVID as well as the severity of its affective symptoms and CFS.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab, Inc., Los Angeles, CA90035, USA
- Cyrex Laboratories, LLC, Phoenix, AZ85034, USA
| | - Abbas F Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu610072, China
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu610072, China
- Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- Research Center, Medical University of Plovdiv, Plovdiv, Bulgaria
- Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul02447, Korea
| |
Collapse
|
2
|
Li Y, Wang H, Zhou J, Wang C. Research progress on the correlation between transforming growth factor- β level and symptoms of depression. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:646-652. [PMID: 37916311 PMCID: PMC10630060 DOI: 10.3724/zdxbyxb-2023-0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/19/2023] [Indexed: 10/08/2023]
Abstract
Transforming growth factor (TGF)-β is a group of cytokines with anti-inflammatory effects in the TGF family, which participates in the development of stress and depression-related mechanisms, and plays roles in the regulation of inflammatory response in depression and the recovery of various cytokine imbalances. The core symptoms of depression is associated with TGF-β level, and the psychological symptoms of depression are related to TGF-β gene polymorphism. Various antidepressants may up-regulate TGF-β level through the complex interaction between neurotransmitters and inflammatory factors, inhibiting inflammatory response and regulating cytokine imbalance to improve depressive symptoms. Studies have shown that recombinant TGF-β1 protein has beneficial effects in mouse depression models, indicating TGF-β1 might be a potential therapeutic target for depression and nasal sprays having the advantage of being fast acting delivery method. This article reviews the research progress on dynamic changes of TGF-β level before and after depression treatment and the application of TGF-β level as an indicator for the improvement of depressive symptoms. We provide ideas for the development of new antidepressants and for the evaluation of the treatment efficacy in depression.
Collapse
Affiliation(s)
- Yanran Li
- Department of Psychiatry, the Second Affiliated Hospital of Xinxiang Medical College, Henan Key Laboratory of Biological Psychiatry, Henan Psychological Aid Cloud Platform and Application Engineering Research Center, Xinxiang 453002, Henan Province, China.
| | - Huiying Wang
- Department of Psychiatry, the Second Affiliated Hospital of Xinxiang Medical College, Henan Key Laboratory of Biological Psychiatry, Henan Psychological Aid Cloud Platform and Application Engineering Research Center, Xinxiang 453002, Henan Province, China
| | - Jiansong Zhou
- Department of Psychiatry, the Second Xiangya Hospital of Central South University, National Clinical Medical Research Center for Ment, Changsha 410011, China.
| | - Changhong Wang
- Department of Psychiatry, the Second Affiliated Hospital of Xinxiang Medical College, Henan Key Laboratory of Biological Psychiatry, Henan Psychological Aid Cloud Platform and Application Engineering Research Center, Xinxiang 453002, Henan Province, China.
| |
Collapse
|
3
|
Valero-Aracama MJ, Zheng F, Alzheimer C. Dorsal-Ventral Gradient of Activin Regulates Strength of GABAergic Inhibition along Longitudinal Axis of Mouse Hippocampus in an Activity-Dependent Fashion. Int J Mol Sci 2023; 24:13145. [PMID: 37685952 PMCID: PMC10487617 DOI: 10.3390/ijms241713145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The functional and neurophysiological distinction between the dorsal and ventral hippocampus affects also GABAergic inhibition. In line with this notion, ventral CA1 pyramidal cells displayed a more dynamic and effective response to inhibitory input compared to their dorsal counterparts. We posit that this difference is effected by the dorsal-ventral gradient of activin A, a member of the transforming growth factor-β family, which is increasingly recognized for its modulatory role in brain regions involved in cognitive functions and affective behavior. Lending credence to this hypothesis, we found that in slices from transgenic mice expressing a dominant-negative mutant of activin receptor IB (dnActRIB), inhibitory transmission was enhanced only in CA1 neurons of the dorsal hippocampus, where the basal activin A level is much higher than in the ventral hippocampus. We next asked how a rise in endogenous activin A would affect GABAergic inhibition along the longitudinal axis of the hippocampus. We performed ex vivo recordings in wild-type and dnActRIB mice after overnight exposure to an enriched environment (EE), which engenders a robust increase in activin A levels in both dorsal and ventral hippocampi. Compared to control mice from standard cages, the behaviorally induced surge in activin A produced a decline in ventral inhibition, an effect that was absent in slices from dnActRIB mice. Underscoring the essential role of activin in the EE-associated modulation of ventral inhibition, this effect was mimicked by acute application of recombinant activin A in control slices. In summary, both genetic and behavioral manipulations of activin receptor signaling affected the dorsal-ventral difference in synaptic inhibition, suggesting that activin A regulates the strength of GABAergic inhibition in a region-specific fashion.
Collapse
|
4
|
Zou ZL, Ye Y, Zhou B, Zhang Y. Identification and characterization of noncoding RNAs-associated competing endogenous RNA networks in major depressive disorder. World J Psychiatry 2023; 13:36-49. [PMID: 36925948 PMCID: PMC10011943 DOI: 10.5498/wjp.v13.i2.36] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/06/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a common and serious mental illness. Many novel genes in MDD have been characterized by high-throughput methods such as microarrays or sequencing. Recently, noncoding RNAs (ncRNAs) were suggested to be involved in the complicated environmental-genetic regulatory network of MDD occurrence; however, the interplay among RNA species, including protein-coding RNAs and ncRNAs, in MDD remains unclear.
AIM To investigate the RNA expression datasets downloaded from a public database and construct a network based on differentially expressed long noncoding RNA (lncRNAs), microRNAs (miRNAs), and mRNAs between MDD and controls.
METHODS Gene expression data were searched in NCBI Gene Expression Omnibus using the search term “major depressive disorder.” Six array datasets from humans were related to the search term: GSE19738, GSE32280, GSE38206, GSE52790, GSE76826, and GSE81152. These datasets were processed for initial assessment and subjected to quality control and differential expression analysis. Differentially expressed lncRNAs, miRNAs, and mRNAs were determined, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed, and protein-protein interaction network was generated. The results were analyzed for their association with MDD.
RESULTS After analysis, 3 miRNAs, 12 lncRNAs, and 33 mRNAs were identified in the competing endogenous RNA network. Two of these miRNAs were earlier shown to be involved in psychiatric disorders, and differentially expressed mRNAs were found to be highly enriched in pathways related to neurogenesis and neuroplasticity as per Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. The expression of hub gene fatty acid 2-hydroxylase was enriched, and the encoded protein was found to be involved in myelin formation, indicating that neurological development and signal transduction are involved in MDD pathogenesis.
CONCLUSION The present study presents candidate ncRNAs involved in the neurogenesis and neuroplasticity pathways related to MDD.
Collapse
Affiliation(s)
- Zhi-Li Zou
- Department of Psychosomatic, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, Chengdu 610072, Sichuan Province, China
| | - Yu Ye
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 611130, Sichuan Province, China
| | - Bo Zhou
- Department of Psychosomatic, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, Chengdu 610072, Sichuan Province, China
| | - Yuan Zhang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu 610072, Sichuan Province, China
| |
Collapse
|
5
|
Morais-Silva G, Campbell RR, Nam H, Basu M, Pagliusi M, Fox ME, Chan CS, Iñiguez SD, Ament S, Cramer N, Marin MT, Lobo MK. Molecular, Circuit, and Stress Response Characterization of Ventral Pallidum Npas1-Neurons. J Neurosci 2023; 43:405-418. [PMID: 36443000 PMCID: PMC9864552 DOI: 10.1523/jneurosci.0971-22.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/31/2022] [Accepted: 11/12/2022] [Indexed: 11/30/2022] Open
Abstract
Altered activity of the ventral pallidum (VP) underlies disrupted motivation in stress and drug exposure. The VP is a very heterogeneous structure composed of many neuron types with distinct physiological properties and projections. Neuronal PAS 1-positive (Npas1+) VP neurons are thought to send projections to brain regions critical for motivational behavior. While Npas1+ neurons have been characterized in the globus pallidus external, there is limited information on these neurons in the VP. To address this limitation, we evaluated the projection targets of the VP Npas1+ neurons and performed RNA-sequencing on ribosome-associated mRNA from VP Npas1+ neurons to determine their molecular identity. Finally, we used a chemogenetic approach to manipulate VP Npas1+ neurons during social defeat stress (SDS) and behavioral tasks related to anxiety and motivation in Npas1-Cre mice. We used a similar approach in females using the chronic witness defeat stress (CWDS). We identified VP Npas1+ projections to the nucleus accumbens, ventral tegmental area, medial and lateral habenula, lateral hypothalamus, thalamus, medial and lateral septum, and periaqueductal gray area. VP Npas1+ neurons displayed distinct translatome representing distinct biological processes. Chemogenetic activation of hM3D(Gq) receptors in VP Npas1+ neurons increased susceptibility to a subthreshold SDS and anxiety-like behavior in the elevated plus maze and open field while the activation of hM4D(Gi) receptors in VP Npas1+ neurons enhanced resilience to chronic SDS and CWDS. Thus, the activity of VP Npas1+ neurons modulates susceptibility to social stressors and anxiety-like behavior. Our studies provide new information on VP Npas1+ neuron circuitry, molecular identity, and their role in stress response.SIGNIFICANCE STATEMENT The ventral pallidum (VP) is a structure connected to both reward-related and aversive brain centers. It is a key brain area that signals the hedonic value of natural rewards. Disruption in the VP underlies altered motivation in stress and substance use disorder. However, VP is a very heterogeneous area with multiple neuron subtypes. This study characterized the projection pattern and molecular signatures of VP Neuronal PAS 1-positive (Npas1+) neurons. We further used tools to alter receptor signaling in VP Npas1+ neurons in stress to demonstrate a role for these neurons in stress behavioral outcomes. Our studies have implications for understanding brain cell type identities and their role in brain disorders, such as depression, a serious disorder that is precipitated by stressful events.
Collapse
Affiliation(s)
- Gessynger Morais-Silva
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, Sao Paulo 14800903, Brazil
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos/Sao Paulo State University, CEP 13565-905, São Carlos/Araraquara, Brazil
| | - Rianne R Campbell
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Hyungwoo Nam
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mahashweta Basu
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Marco Pagliusi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Department of Structural and Functional Biology, State University of Campinas, SP-13083-872, Campinas, Brazil
| | - Megan E Fox
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - C Savio Chan
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Sergio D Iñiguez
- Department of Psychology, University of Texas at El Paso, El Paso, Texas 79902
| | - Seth Ament
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Nathan Cramer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Marcelo Tadeu Marin
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, Sao Paulo 14800903, Brazil
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos/Sao Paulo State University, CEP 13565-905, São Carlos/Araraquara, Brazil
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
6
|
Dahlmanns M, Dahlmanns JK, Schmidt CC, Valero-Aracama MJ, Zheng F, Alzheimer C. Environmental enrichment recruits activin A to recalibrate neural activity in mouse hippocampus. Cereb Cortex 2023; 33:663-675. [PMID: 35257169 DOI: 10.1093/cercor/bhac092] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 02/03/2023] Open
Abstract
The TGF-β family member activin A modulates neural underpinnings of cognitive and affective functions in an activity-dependent fashion. We have previously shown that exploration of a novel and enriched environment (EE) strongly enhanced activin signaling. Whereas the many beneficial effects of EE are amply documented, the underlying mechanisms remain largely elusive. Here, we examined the hypothesis that EE recruits activin to regulate synaptic plasticity in a coordinated, cognition-promoting manner. Elevated activin levels after EE enhanced CA1 pyramidal cell excitability, facilitated synaptic transmission, and promoted long-term potentiation. These EE-induced changes were largely absent in mice expressing a dominant-negative mutant of activin receptor IB. We then interrogated the impact of activin on network oscillations and functional connectivity, using high-speed Ca 2+ imaging to study spike routing within networks formed by dissociated primary hippocampal cultures. Activin facilitated Ca2+ signaling, enhanced the network strength, and shortened the weighted characteristic path length. In the slice preparation, activin promoted theta oscillations during cholinergic stimulation. Thus, we advance activin as an activity-dependent and very early molecular effector that translates behavioral stimuli experienced during EE exposure into a set of synchronized changes in neuronal excitability, synaptic plasticity, and network activity that are all tuned to improve cognitive functions.
Collapse
Affiliation(s)
- Marc Dahlmanns
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jana Katharina Dahlmanns
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Carla C Schmidt
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Maria Jesus Valero-Aracama
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
7
|
Zheng F, Valero-Aracama MJ, Schaefer N, Alzheimer C. Activin A Reduces GIRK Current to Excite Dentate Gyrus Granule Cells. Front Cell Neurosci 2022; 16:920388. [PMID: 35711474 PMCID: PMC9197229 DOI: 10.3389/fncel.2022.920388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Activin A, a member of the TGF-β family, is recognized as a multifunctional protein in the adult brain with a particular impact on neuronal circuits associated with cognitive and affective functions. Activin receptor signaling in mouse hippocampus is strongly enhanced by the exploration of an enriched environment (EE), a behavioral paradigm known to improve performance in learning and memory tasks and to ameliorate depression-like behaviors. To interrogate the relationship between EE, activin signaling, and cellular excitability in the hippocampus, we performed ex vivo whole-cell recordings from dentate gyrus (DG) granule cells (GCs) of wild type mice and transgenic mice expressing a dominant-negative mutant of activin receptor IB (dnActRIB), which disrupts activin signaling in a forebrain-specific fashion. We found that, after overnight EE housing, GC excitability was strongly enhanced in an activin-dependent fashion. Moreover, the effect of EE on GC firing was mimicked by pre-treatment of hippocampal slices from control mice with recombinant activin A for several hours. The excitatory effect of activin A was preserved when canonical SMAD-dependent signaling was pharmacologically suppressed but was blocked by inhibitors of ERK-MAPK and PKA signaling. The involvement of a non-genomic signaling cascade was supported by the fact that the excitatory effect of activin A was already achieved within minutes of application. With respect to the ionic mechanism underlying the increase in intrinsic excitability, voltage-clamp recordings revealed that activin A induced an apparent inward current, which resulted from the suppression of a standing G protein-gated inwardly rectifying K+ (GIRK) current. The link between EE, enhanced activin signaling, and inhibition of GIRK current was strengthened by the following findings: (i) The specific GIRK channel blocker tertiapin Q (TQ) occluded the characteristic electrophysiological effects of activin A in both current- and voltage-clamp recordings. (ii) The outward current evoked by the GIRK channel activator adenosine was significantly reduced by preceding EE exploration as well as by recombinant activin A in control slices. In conclusion, our study identifies GIRK current suppression via non-canonical activin signaling as a mechanism that might at least in part contribute to the beneficial effects of EE on cognitive performance and affective behavior.
Collapse
Affiliation(s)
- Fang Zheng
- Institute of Physiology
and Pathophysiology, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Christian Alzheimer Fang Zheng
| | - Maria Jesus Valero-Aracama
- Institute of Physiology
and Pathophysiology, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Christian Alzheimer
- Institute of Physiology
and Pathophysiology, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Christian Alzheimer Fang Zheng
| |
Collapse
|
8
|
Schmidt CC, Zheng F, Alzheimer C. Activin A regulates the excitability of hippocampal mossy cells. Hippocampus 2022; 32:401-410. [PMID: 35301773 DOI: 10.1002/hipo.23415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/15/2022] [Accepted: 03/11/2022] [Indexed: 12/16/2022]
Abstract
Mossy cells (MCs) in the hilus of the dentate gyrus (DG) receive increasing attention as a major player controlling information processing in the DG network. Furthermore, disturbed MC activity has been implicated in widespread neuropsychiatric disorders such as epilepsy and major depression. Using whole-cell patch-clamp recordings from MCs in acute hippocampal slices from wild type and transgenic mice, we demonstrate that activin, a member of the transforming growth factor-β (TGF-β) family, has a strong neuromodulatory effect on MC activity. Disruption of activin receptor signaling reduced MC firing, dampened their excitatory input and augmented their inhibitory input. By contrast, acute application of recombinant activin A strongly increased MC activity and promoted excitatory synaptic drive. Notably, similar changes of MC activity have been observed in a rodent model of depression and after antidepressant drug therapy, respectively. Given that a rise in activin signaling particularly in the DG has been proposed as a mechanism of antidepressant action, our data suggest that the effect of activin on MC excitability might make a considerable contribution in this regard.
Collapse
Affiliation(s)
- Carla C Schmidt
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Ho CH, Paolantoni C, Bawankar P, Tang Z, Brown S, Roignant J, Treisman JE. An exon junction complex-independent function of Barentsz in neuromuscular synapse growth. EMBO Rep 2022; 23:e53231. [PMID: 34726300 PMCID: PMC8728599 DOI: 10.15252/embr.202153231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
The exon junction complex controls the translation, degradation, and localization of spliced mRNAs, and three of its core subunits also play a role in splicing. Here, we show that a fourth subunit, Barentsz, has distinct functions within and separate from the exon junction complex in Drosophila neuromuscular development. The distribution of mitochondria in larval muscles requires Barentsz as well as other exon junction complex subunits and is not rescued by a Barentsz transgene in which residues required for binding to the core subunit eIF4AIII are mutated. In contrast, interactions with the exon junction complex are not required for Barentsz to promote the growth of neuromuscular synapses. We find that the Activin ligand Dawdle shows reduced expression in barentsz mutants and acts downstream of Barentsz to control synapse growth. Both barentsz and dawdle are required in motor neurons, muscles, and glia for normal synapse growth, and exogenous Dawdle can rescue synapse growth in the absence of barentsz. These results identify a biological function for Barentsz that is independent of the exon junction complex.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Skirball Institute for Biomolecular Medicine and Department of Cell BiologyNYU School of MedicineNew YorkNYUSA
| | - Chiara Paolantoni
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
| | - Praveen Bawankar
- Institute of Pharmaceutical and Biomedical SciencesJohannes Gutenberg‐University MainzMainzGermany
| | - Zuojian Tang
- Center for Health Informatics and BioinformaticsNYU Langone Medical CenterNew YorkNYUSA
- Present address:
Computational Biology at Ridgefield US, Global Computational Biology and Digital ScienceBoehringer IngelheimRidgefieldCTUSA
| | - Stuart Brown
- Center for Health Informatics and BioinformaticsNYU Langone Medical CenterNew YorkNYUSA
- Present address:
ExxonMobil Corporate Strategic ResearchAnnandaleNJUSA
| | - Jean‐Yves Roignant
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
- Institute of Pharmaceutical and Biomedical SciencesJohannes Gutenberg‐University MainzMainzGermany
| | - Jessica E Treisman
- Skirball Institute for Biomolecular Medicine and Department of Cell BiologyNYU School of MedicineNew YorkNYUSA
| |
Collapse
|
10
|
Neuroadaptations and TGF-β signaling: emerging role in models of neuropsychiatric disorders. Mol Psychiatry 2022; 27:296-306. [PMID: 34131268 PMCID: PMC8671568 DOI: 10.1038/s41380-021-01186-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
Neuropsychiatric diseases are manifested by maladaptive behavioral plasticity. Despite the greater understanding of the neuroplasticity underlying behavioral adaptations, pinpointing precise cellular mediators has remained elusive. This has stymied the development of pharmacological interventions to combat these disorders both at the level of progression and relapse. With increased knowledge on the putative role of the transforming growth factor (TGF- β) family of proteins in mediating diverse neuroadaptations, the influence of TGF-β signaling in regulating maladaptive cellular and behavioral plasticity underlying neuropsychiatric disorders is being increasingly elucidated. The current review is focused on what is currently known about the TGF-β signaling in the central nervous system in mediating cellular and behavioral plasticity related to neuropsychiatric manifestations.
Collapse
|
11
|
Bu T, Qiao Z, Wang W, Yang X, Zhou J, Chen L, Yang J, Xu J, Ji Y, Wang Y, Zhang W, Yang Y, Qiu X, Yu Y. Diagnostic Biomarker Hsa_circ_0126218 and Functioning Prediction in Peripheral Blood Monocular Cells of Female Patients With Major Depressive Disorder. Front Cell Dev Biol 2021; 9:651803. [PMID: 34095115 PMCID: PMC8174117 DOI: 10.3389/fcell.2021.651803] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Introduction Although major depressive diroder (MDD) has brought huge burden and challenges to society globally, effective and accurate diagnoses and treatments remain inadequate. The pathogenesis that for women are more likely to suffer from depression than men needs to be excavated as well. The function of circRNAs in pathological process of depression has not been widely investigated. This study aims to explore potential diagnostic biomarker circRNA of female patients with MDD and to investigate its role in pathogenesis. Methods First, an expression profile of circRNAs in the peripheral blood monocular cells of MDD patients and healthy peripherals were established based on high-throughput sequencing analysis. In addition, the top 10 differentially expressed circRNAs were quantified by quantitative real-time PCR to explore diagnostic biomarkers. To further investigate the function of biomarkers in the pathogenesis of MDD, bioinformatics analysis on downstream target genes of the biomarkers was carried out. Results There is a mass of dysregulated circRNAs in PBMCs between female MDD patients and healthy controls. Among the top 10 differentially expressed circRNAs, hsa_circ_0126218 is more feasible as a diagnostic biomarker. The expression level of hsa_circ_0126218 displayed upregulation in patients with MDD and the area under the operating characteristic curve of hsa_circ_0126218 was 0.801 (95% CI 0.7226–0.8791, p < 0.0001). To explain the competing endogenous RNA role of hsa_circ_0126218 in the pathogenesis of female MDD, a hsa_circ_0126218-miRNA-mRNA network was established. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses stated that some of the enriched pathways downstream of hsa_circ_0126218 are closely related to MDD. Moreover, we established a protein-protein network to further screen out the hub genes (PIK3CA, PTEN, MAPK1, CDC42, Lyn, YES1, EPHB2, SMAD2, STAT1, and ILK). The function of hsa_circ_0126218 was refined by constructing a verified circRNA-predicted miRNA-hub gene subnetwork. Conclusion hsa_circ_0126218 can be considered as a new female MDD biomarker, and the pathogenesis of female MDD by the downstream regulation of hsa_circ_0126218 has been predicted. These findings may help further improve the early detection, effective diagnosis, convenient monitoring of complications, precise treatment, and timely recurrence prevention of depression.
Collapse
Affiliation(s)
- Tianyi Bu
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Zhengxue Qiao
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Wenbo Wang
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Xiuxian Yang
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Jiawei Zhou
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Lu Chen
- Department of Endocrinology, Peking Union Medical College Hospital, Beijing, China
| | - Jiarun Yang
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Jia Xu
- Psychotherapy Department, The First Psychiatric Hospital of Harbin, Harbin, China
| | - Yanping Ji
- Department of Nursing, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yini Wang
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Wenxin Zhang
- Medical Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanjie Yang
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Xiaohui Qiu
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| | - Yunmiao Yu
- Psychology and Health Management Center, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
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
|
13
|
Zou T, Zhang J, Liu Y, Zhang Y, Sugimoto K, Mei C. Antidepressant-Like Effect of Geniposide in Mice Exposed to a Chronic Mild Stress Involves the microRNA-298-5p-Mediated Nox1. Front Mol Neurosci 2021; 13:131. [PMID: 33613190 PMCID: PMC7886707 DOI: 10.3389/fnmol.2020.00131] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
Depression is a common mental disorder that presents a considerable challenge for public health. The natural product geniposide has neuroprotective effects on depression, but the underlying mechanism behind these effects had remained undefined. The present study was designed to investigate the role of microRNAs (miRs) in this mechanism. It studied mice with depression-like behavior established by exposure to chronic unpredictable mild stress (CUMS) for 2 months. The CUMS mice were intragastrically fed with geniposide at a dose of 10 ml/kg daily for two consecutive weeks. We monitored the depression-like behaviors of the CUMS mice by the forced swimming test (FST) and tail suspension test (TST). Then, we measured the cerebral expression of miR-298-5p and NADPH oxidase 1 (Nox1) mRNA in the CUMS mice by the RT-qPCR. The targeting relationship between miR-298-5p and Nox1 was evaluated by dual-luciferase reporter gene assay. The concentrations of adenosine triphosphate (ATP) and reactive oxygen species (ROS) were determined by the CellTiter-Glo® and flow cytometry, respectively. The mitochondrial membrane potential (MMP) was detected using JC-1 staining. Moreover, the expression of inflammatory cytokines (TNF-α, IL-1β, IL-6, and TGF-β) was determined by ELISA, RT-qPCR, and western blot analysis. We found that miR-298-5p was poorly-expressed while Nox1 was highly-expressed in the brain tissues of the CUMS-induced mice. Intriguingly, Geniposide treatment reversed the behavioral abnormalities of CUMS mice, including shortened immobility time. Geniposide inhibited the Nox1 expression by increasing miR-298-5p levels. There were increased ATP content and MMP and reduced contents of ROS and inflammatory cytokines in the CUMS mice receiving geniposide treatment. Hence, this study revealed an antidepressant effect of geniposide on CUMS-induced depression-like behavior in mice by down-regulating the miR-298-5p-targeted Nox1. This highlights a novel candidate target for the treatment of depression.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of Encephalopathy, Heilongjiang Academy of Chinese Medical Sciences, Harbin, China
| | - Jielin Zhang
- Department of Dermatology, Heilongjiang Provincial Hospital Affiliated to Harbin Institute of Technology, Harbin, China
| | - Yongxiu Liu
- Department of Encephalopathy, Heilongjiang Academy of Chinese Medical Sciences, Harbin, China
| | - Yiming Zhang
- Department of Encephalopathy, Heilongjiang Academy of Chinese Medical Sciences, Harbin, China
| | - Kazuo Sugimoto
- Department of Neurology, Dongzhimen Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Mei
- Department of Encephalopathy, Heilongjiang Academy of Chinese Medical Sciences, Harbin, China
| |
Collapse
|
14
|
Dentate gyrus activin signaling mediates the antidepressant response. Transl Psychiatry 2021; 11:7. [PMID: 33414389 PMCID: PMC7791138 DOI: 10.1038/s41398-020-01156-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Antidepressants that target monoaminergic systems, such as selective serotonin reuptake inhibitors (SSRIs), are widely used to treat neuropsychiatric disorders including major depressive disorder, several anxiety disorders, and obsessive-compulsive disorder. However, these treatments are not ideal because only a subset of patients achieve remission. The reasons why some individuals remit to antidepressant treatments while others do not are unknown. Here, we developed a paradigm to assess antidepressant treatment resistance in mice. Exposure of male C57BL/6J mice to either chronic corticosterone administration or chronic social defeat stress induces maladaptive affective behaviors. Subsequent chronic treatment with the SSRI fluoxetine reverses these maladaptive affective behavioral changes in some, but not all, of the mice, permitting stratification into persistent responders and non-responders to fluoxetine. We found several differences in expression of Activin signaling-related genes between responders and non-responders in the dentate gyrus (DG), a region that is critical for the beneficial behavioral effects of fluoxetine. Enhancement of Activin signaling in the DG converted behavioral non-responders into responders to fluoxetine treatment more effectively than commonly used second-line antidepressant treatments, while inhibition of Activin signaling in the DG converted responders into non-responders. Taken together, these results demonstrate that the behavioral response to fluoxetine can be bidirectionally modified via targeted manipulations of the DG and suggest that molecular- and neural circuit-based modulations of DG may provide a new therapeutic avenue for more effective antidepressant treatments.
Collapse
|
15
|
Zhao M, Chen L, Qiao Z, Zhou J, Zhang T, Zhang W, Ke S, Zhao X, Qiu X, Song X, Zhao E, Pan H, Yang Y, Yang X. Association Between FoxO1, A2M, and TGF-β1, Environmental Factors, and Major Depressive Disorder. Front Psychiatry 2020; 11:675. [PMID: 32792993 PMCID: PMC7394695 DOI: 10.3389/fpsyt.2020.00675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 06/29/2020] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Investigations of gene-environment (G×E) interactions in major depressive disorder (MDD) have been limited to hypothesis testing of candidate genes while poly-gene-environmental causation has not been adequately address. To this end, the present study analyzed the association between three candidate genes, two environmental factors, and MDD using a hypothesis-free testing approach. METHODS A logistic regression model was used to analyze interaction effects; a hierarchical regression model was used to evaluate the effects of different genotypes and the dose-response effects of the environment; genetic risk score (GRS) was used to estimate the cumulative contribution of genetic factors to MDD; and protein-protein interaction (PPI) analyses were carried out to evaluate the relationship between candidate genes and top MDD susceptibility genes. RESULTS Allelic association analyses revealed significant effects of the interaction between the candidate genes Forkhead box (Fox)O1, α2-macroglobulin (A2M), and transforming growth factor (TGF)-β1 genes and the environment on MDD. Gene-gene (G×G) and gene-gene-environment (G×G×E) interactions in MDD were also included in the model. Hierarchical regression analysis showed that the effect of environmental factors on MDD was greater in homozygous than in heterozygous mutant genotypes of the FoxO1 and TGF-β1 genes; a dose-response effect between environment and MDD on genotypes was also included in this model. Haplotype analyses revealed significant global and individual effects of haplotypes on MDD in the whole sample as well as in subgroups. There was a significant association between GRS and MDD (P = 0.029) and a GRS and environment interaction effect on MDD (P = 0.009). Candidate and top susceptibility genes were connected in PPI networks. CONCLUSIONS FoxO1, A2M, and TGF-β1 interact with environmental factors and with each other in MDD. Multi-factorial G×E interactions may be responsible for a higher explained variance and may be associated with causal factors and mechanisms that could inform new diagnosis and therapeutic strategies, which can contribute to the personalized medicine of MDD.
Collapse
Affiliation(s)
- Mingzhe Zhao
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Lu Chen
- Department of Endocrinology, Peking Union Medical College Hospital, Beijing, China
| | - Zhengxue Qiao
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Jiawei Zhou
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Tianyu Zhang
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Wenxin Zhang
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Siyuan Ke
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Xiaoyun Zhao
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Xiaohui Qiu
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Xuejia Song
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Erying Zhao
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Hui Pan
- Department of Endocrinology, Peking Union Medical College Hospital, Beijing, China
| | - Yanjie Yang
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| | - Xiuxian Yang
- Psychology Department, Public Health Institute, Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Nava N, Treccani G, Alabsi A, Kaastrup Mueller H, Elfving B, Popoli M, Wegener G, Nyengaard JR. Temporal Dynamics of Acute Stress-Induced Dendritic Remodeling in Medial Prefrontal Cortex and the Protective Effect of Desipramine. Cereb Cortex 2018; 27:694-705. [PMID: 26523035 DOI: 10.1093/cercor/bhv254] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Stressful events are associated with increased risk of mood disorders. Volumetric reductions have been reported in brain areas critical for the stress response, such as medial prefrontal cortex (mPFC), and dendritic remodeling has been proposed as an underlying factor. Here, we investigated the time-dependent effects of acute stress on dendritic remodeling within the prelimbic (PL) region of the PFC, and whether treatment with the antidepressant desipramine (DMI) may interfere. Rodents were subjected to foot-shock stress: dendritic length and spine density were analyzed 1 day, 7 days, and 14 days after stress. Acute stress produced increased spine density and decreased cofilin phosphorylation at 1 day, paralleled with dendritic retraction. An overall shift in spine population was observed at 1 day, resulting in a stress-induced increase in small spines. Significant atrophy of apical dendrites was observed at 1 day, which was prevented by chronic DMI, and at 14 days after stress exposure. Chronic DMI resulted in dendritic elaboration at 7 days but did not prevent the effects of FS-stress. Collectively, these data demonstrate that 1) acute stressors may induce rapid and sustained changes of PL neurons; and 2) chronic DMI may protect neurons from rapid stress-induced synaptic changes.
Collapse
Affiliation(s)
- Nicoletta Nava
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, Aarhus C 8000, Denmark.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark
| | - Giulia Treccani
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark.,Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Milan 20133, Italy
| | - Abdelrahman Alabsi
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging,Aarhus University Hospital, Aarhus C 8000, Denmark
| | - Heidi Kaastrup Mueller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Milan 20133, Italy
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark.,Pharmaceutical Research Center of Excellence, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Jens Randel Nyengaard
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging,Aarhus University Hospital, Aarhus C 8000, Denmark
| |
Collapse
|
17
|
Trojan E, Ślusarczyk J, Chamera K, Kotarska K, Głombik K, Kubera M, Basta-Kaim A. The Modulatory Properties of Chronic Antidepressant Drugs Treatment on the Brain Chemokine - Chemokine Receptor Network: A Molecular Study in an Animal Model of Depression. Front Pharmacol 2017; 8:779. [PMID: 29163165 PMCID: PMC5671972 DOI: 10.3389/fphar.2017.00779] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/16/2017] [Indexed: 12/26/2022] Open
Abstract
An increasing number of studies indicate that the chemokine system may be the third major communication system of the brain. Therefore, the role of the chemokine system in the development of brain disorders, including depression, has been recently proposed. However, little is known about the impact of the administration of various antidepressant drugs on the brain chemokine - chemokine receptor axis. In the present study, we used an animal model of depression based on the prenatal stress procedure. We determined whether chronic treatment with tianeptine, venlafaxine, or fluoxetine influenced the evoked by prenatal stress procedure changes in the mRNA and protein levels of the homeostatic chemokines, CXCL12 (SDF-1α), CX3CL1 (fractalkine) and their receptors, in the hippocampus and frontal cortex. Moreover, the impact of mentioned antidepressants on the TGF-β, a molecular pathway related to fractalkine receptor (CX3CR1), was explored. We found that prenatal stress caused anxiety and depressive-like disturbances in adult offspring rats, which were normalized by chronic antidepressant treatment. Furthermore, we showed the stress-evoked CXCL12 upregulation while CXCR4 downregulation in hippocampus and frontal cortex. CXCR7 expression was enhanced in frontal cortex but not hippocampus. Furthermore, the levels of CX3CL1 and CX3CR1 were diminished by prenatal stress in the both examined brain areas. The mentioned changes were normalized with various potency by chronic administration of tested antidepressants. All drugs in hippocampus, while tianeptine and venlafaxine in frontal cortex normalized the CXCL12 level in prenatally stressed offspring. Moreover, in hippocampus only fluoxetine enhanced CXCR4 level, while fluoxetine and tianeptine diminished CXCR7 level in frontal cortex. Additionally, the diminished by prenatal stress levels of CX3CL1 and CX3CR1 in the both examined brain areas were normalized by chronic tianeptine and partially fluoxetine administration. Tianeptine modulate also brain TGF-β signaling in the prenatal stress-induced animal model of depression. Our results provide new evidence that not only prenatal stress-induced behavioral disturbances but also changes of CXCL12 and their receptor and at less extend in CX3CL1-CX3CR1 expression may be normalized by chronic antidepressant drug treatment. In particular, the effect on the CXCL12 and their CXCR4 and CXCR7 receptors requires additional studies to elucidate the possible biological consequences.
Collapse
Affiliation(s)
- Ewa Trojan
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Joanna Ślusarczyk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Chamera
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Kotarska
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Głombik
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Marta Kubera
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
18
|
Abstract
Depression is a polygenic and highly complex psychiatric disorder that remains a major burden on society. Antidepressants, such as selective serotonin reuptake inhibitors (SSRIs), are some of the most commonly prescribed drugs worldwide. In this review, we will discuss the evidence that links serotonin and serotonin receptors to the etiology of depression and the mechanisms underlying response to antidepressant treatment. We will then revisit the role of serotonin in three distinct hypotheses that have been proposed over the last several decades to explain the pathophysiology of depression: the monoamine, neurotrophic, and neurogenic hypotheses. Finally, we will discuss how recent studies into serotonin receptors have implicated specific neural circuitry in mediating the antidepressant response, with a focus being placed on the hippocampus.
Collapse
Affiliation(s)
- Christine N Yohn
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA
| | - Mark M Gergues
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA
| | - Benjamin Adam Samuels
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA.
| |
Collapse
|
19
|
Link AS, Zheng F, Alzheimer C. Activin Signaling in the Pathogenesis and Therapy of Neuropsychiatric Diseases. Front Mol Neurosci 2016; 9:32. [PMID: 27242425 PMCID: PMC4861723 DOI: 10.3389/fnmol.2016.00032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/25/2016] [Indexed: 01/29/2023] Open
Abstract
Activins are members of the transforming growth factor β (TGFβ) family and serve as multifunctional regulatory proteins in many tissues and organs. In the brain, activin A, which is formed by two disulfide-linked βA subunits, is recognized as the predominant player in activin signaling. Over the last years, considerable progress has been made in elucidating novel and unexpected functions of activin in the normal and diseased brain and in deciphering the underlying molecular mechanisms. Initially identified as a neurotrophic and protective factor during development and in several forms of acute injury, the scope of effects of activin A in the adult central nervous system (CNS) has been considerably broadened by now. Here, we will highlight recent findings that bear significance for a better understanding of the pathogenesis of various neuropsychiatric diseases and might hold promise for novel therapeutic strategies. While the basal level of activin A in the adult brain is low, significant short-term up-regulation occurs in response to increased neuronal activity. In fact, brief exposure to an enriched environment (EE) is already sufficient to considerably strengthen activin signaling. Enhancement of this pathway tunes the performance of glutamatergic and GABAergic synapses in a fashion that impacts on cognitive functions and affective behavior, counteracts death-inducing signals through extrasynaptic NMDA receptors (NMDARs), and stimulates adult neurogenesis in the hippocampus. We will discuss how impaired activin signaling is involved in anxiety disorders, depression, drug dependence, and neurodegenerative diseases such as Alzheimer’s and Parkinson’s, and how reinforcement of activin signaling might be exploited for therapeutic interventions.
Collapse
Affiliation(s)
- Andrea S Link
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany
| | - Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany
| |
Collapse
|
20
|
Expression and role of the TGF-β family in glial cells infected with Borna disease virus. Microbes Infect 2016; 18:128-36. [DOI: 10.1016/j.micinf.2015.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/09/2015] [Accepted: 10/09/2015] [Indexed: 12/21/2022]
|
21
|
Link AS, Kurinna S, Havlicek S, Lehnert S, Reichel M, Kornhuber J, Winner B, Huth T, Zheng F, Werner S, Alzheimer C. Kdm6b and Pmepa1 as Targets of Bioelectrically and Behaviorally Induced Activin A Signaling. Mol Neurobiol 2015. [PMID: 26215835 DOI: 10.1007/s12035-015-9363-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The transforming growth factor-β (TGF-β) family member activin A exerts multiple neurotrophic and protective effects in the brain. Activin also modulates cognitive functions and affective behavior and is a presumed target of antidepressant therapy. Despite its important role in the injured and intact brain, the mechanisms underlying activin effects in the CNS are still largely unknown. Our goal was to identify the first target genes of activin signaling in the hippocampus in vivo. Electroconvulsive seizures, a rodent model of electroconvulsive therapy in humans, were applied to C57BL/6J mice to elicit a strong increase in activin A signaling. Chromatin immunoprecipitation experiments with hippocampal lysates subsequently revealed that binding of SMAD2/3, the intracellular effectors of activin signaling, was significantly enriched at the Pmepa1 gene, which encodes a negative feedback regulator of TGF-β signaling in cancer cells, and at the Kdm6b gene, which encodes an epigenetic regulator promoting transcriptional plasticity. Underlining the significance of these findings, activin treatment also induced PMEPA1 and KDM6B expression in human forebrain neurons generated from embryonic stem cells suggesting interspecies conservation of activin effects in mammalian neurons. Importantly, physiological stimuli such as provided by environmental enrichment proved already sufficient to engender a rapid and significant induction of activin signaling concomitant with an upregulation of Pmepa1 and Kdm6b expression. Taken together, our study identified the first target genes of activin signaling in the brain. With the induction of Kdm6b expression, activin is likely to gain impact on a presumed epigenetic regulator of activity-dependent neuronal plasticity.
Collapse
Affiliation(s)
- Andrea S Link
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Germany
| | - Svitlana Kurinna
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Steven Havlicek
- IZKF Junior Research Group and BMBF Research Group Neuroscience, IZKF, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glückstr. 6, 91054, Erlangen, Germany
- Present address: Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, 60 Biopolis Street, 138672, Singapore, Singapore
| | - Sandra Lehnert
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Beate Winner
- IZKF Junior Research Group and BMBF Research Group Neuroscience, IZKF, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glückstr. 6, 91054, Erlangen, Germany
| | - Tobias Huth
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Germany
| | - Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Germany
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Germany.
| |
Collapse
|
22
|
Gancarz AM, Wang ZJ, Schroeder GL, Damez-Werno D, Braunscheidel K, Mueller LE, Humby MS, Caccamise A, Martin JA, Dietz KC, Neve RL, Dietz DM. Activin receptor signaling regulates cocaine-primed behavioral and morphological plasticity. Nat Neurosci 2015; 18:959-61. [PMID: 26030849 PMCID: PMC4599345 DOI: 10.1038/nn.4036] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/29/2015] [Indexed: 02/06/2023]
Abstract
Activin receptor signaling, including the transcription factor Smad3, was upregulated in the rat nucleus accumbens (NAc) shell following withdrawal from cocaine. Direct genetic and pharmacological manipulations of this pathway bidirectionally altered cocaine seeking while governing morphological plasticity in NAc neurons. Thus, Activin/Smad3 signaling is induced following withdrawal from cocaine, and such regulation may be a key molecular mechanism underlying behavioral and cellular plasticity in the brain following cocaine self-administration.
Collapse
Affiliation(s)
- Amy M. Gancarz
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Zi-Jun Wang
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Gabrielle L. Schroeder
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Diane Damez-Werno
- Department of Neuroscience at Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kevin Braunscheidel
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Lauren E. Mueller
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Monica S. Humby
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Aaron Caccamise
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Jennifer A. Martin
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | - Karen C. Dietz
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| | | | - David M. Dietz
- Department of Pharmacology and Toxicology; Research Institute on Addictions; Program in Neuroscience, State University of New York at Buffalo, Buffalo, NY
| |
Collapse
|
23
|
Yamasaki A, Kasai A, Toi A, Kurita M, Kimoto S, Hayata-Takano A, Nakazawa T, Nagayasu K, Shintani N, Hashimoto R, Ito A, Meltzer HY, Ago Y, Waschek JA, Onaka Y, Matsuda T, Baba A, Hashimoto H. Identification of the role of bone morphogenetic protein (BMP) and transforming growth factor-β (TGF-β) signaling in the trajectory of serotonergic differentiation in a rapid assay in mouse embryonic stem cells in vitro. J Neurochem 2015; 132:418-28. [PMID: 25421849 DOI: 10.1111/jnc.12999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/18/2014] [Accepted: 11/15/2014] [Indexed: 12/25/2022]
Abstract
The mechanism by which extracellular molecules control serotonergic cell fate remains elusive. Recently, we showed that noggin, which inactivates bone morphogenetic proteins (BMPs), induces serotonergic differentiation of mouse embryonic (ES) and induced pluripotent stem cells with coordinated gene expression along the serotonergic lineage. Here, we created a rapid assay for serotonergic induction by generating knock-in ES cells expressing a naturally secreted Gaussia luciferase driven by the enhancer of Pet-1/Fev, a landmark of serotonergic differentiation. Using these cells, we performed candidate-based screening and identified BMP type I receptor kinase inhibitors LDN-193189 and DMH1 as activators of luciferase. LDN-193189 induced ES cells to express the genes encoding Pet-1, tryptophan hydroxylase 2, and the serotonin transporter, and increased serotonin release without altering dopamine release. In contrast, TGF-β receptor inhibitor SB-431542 selectively inhibited serotonergic differentiation, without changing overall neuronal differentiation. LDN-193189 inhibited expression of the BMP signaling target gene Id, and induced the TGF-β target gene Lefty, whereas the opposite effect was observed with SB-431542. This study thus provides a new tool to investigate serotonergic differentiation and suggests that inhibition of BMP type I receptors and concomitant activation of TGF-β receptor signaling are implicated in serotonergic differentiation. Candidate-based screening for serotonergic induction using a rapid assay in mouse embryonic stem cells revealed that the bone morphogenetic protein (BMP) type I receptor kinase inhibitors selectively induce serotonergic differentiation, whereas the TGF-β receptor inhibitor SB-431542 inhibits the differentiation. These results suggest that inhibition of BMP type I receptors and concomitant activation of transforming growth factor-β (TGF-β) receptor signaling are involved in the early trajectory of serotonergic differentiation.
Collapse
Affiliation(s)
- Atsushi Yamasaki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Levinstein MR, Samuels BA. Mechanisms underlying the antidepressant response and treatment resistance. Front Behav Neurosci 2014; 8:208. [PMID: 25018708 PMCID: PMC4073308 DOI: 10.3389/fnbeh.2014.00208] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/22/2014] [Indexed: 12/28/2022] Open
Abstract
Depression is a complex and heterogeneous disorder affecting millions of Americans. There are several different medications and other treatments that are available and effective for many patients with depression. However, a substantial percentage of patients fail to achieve remission with these currently available interventions, and relapse rates are high. Therefore, it is necessary to determine both the mechanisms underlying the antidepressant response and the differences between responders and non-responders to treatment. Delineation of these mechanisms largely relies on experiments that utilize animal models. Therefore, this review provides an overview of the various mouse models that are currently used to assess the antidepressant response, such as chronic mild stress, social defeat, and chronic corticosterone. We discuss how these mouse models can be used to advance our understanding of the differences between responders and non-responders to antidepressant treatment. We also provide an overview of experimental treatment modalities that are used for treatment-resistant depression, such as deep brain stimulation and ketamine administration. We will then review the various genetic polymorphisms and transgenic mice that display resistance to antidepressant treatment. Finally, we synthesize the published data to describe a potential neural circuit underlying the antidepressant response and treatment resistance.
Collapse
Affiliation(s)
- Marjorie R Levinstein
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, Research Foundation for Mental Hygiene, Inc. New York, NY, USA
| | - Benjamin A Samuels
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, Research Foundation for Mental Hygiene, Inc. New York, NY, USA
| |
Collapse
|
25
|
Hasegawa Y, Mukai H, Asashima M, Hojo Y, Ikeda M, Komatsuzaki Y, Ooishi Y, Kawato S. Acute modulation of synaptic plasticity of pyramidal neurons by activin in adult hippocampus. Front Neural Circuits 2014; 8:56. [PMID: 24917791 PMCID: PMC4040441 DOI: 10.3389/fncir.2014.00056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/11/2014] [Indexed: 11/25/2022] Open
Abstract
Activin A is known as a neuroprotective factor produced upon acute excitotoxic injury of the hippocampus (in pathological states). We attempt to reveal the role of activin as a neuromodulator in the adult male hippocampus under physiological conditions (in healthy states), which remains largely unknown. We showed endogenous/basal expression of activin in the hippocampal neurons. Localization of activin receptors in dendritic spines (=postsynapses) was demonstrated by immunoelectron microscopy. The incubation of hippocampal acute slices with activin A (10 ng/mL, 0.4 nM) for 2 h altered the density and morphology of spines in CA1 pyramidal neurons. The total spine density increased by 1.2-fold upon activin treatments. Activin selectively increased the density of large-head spines, without affecting middle-head and small-head spines. Blocking Erk/MAPK, PKA, or PKC prevented the activin-induced spinogenesis by reducing the density of large-head spines, independent of Smad-induced gene transcription which usually takes more than several hours. Incubation of acute slices with activin for 2 h induced the moderate early long-term potentiation (moderate LTP) upon weak theta burst stimuli. This moderate LTP induction was blocked by follistatin, MAPK inhibitor (PD98059) and inhibitor of NR2B subunit of NMDA receptors (Ro25-6981). It should be noted that the weak theta burst stimuli alone cannot induce moderate LTP. These results suggest that MAPK-induced phosphorylation of NMDA receptors (including NR2B) may play an important role for activin-induced moderate LTP. Taken together, the current results reveal interesting physiological roles of endogenous activin as a rapid synaptic modulator in the adult hippocampus.
Collapse
Affiliation(s)
- Yoshitaka Hasegawa
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Hideo Mukai
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan ; Bioinformatics Project (BIRD), Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Core Research for Evolutional Science and Technology Project of Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Department of Computer Science, School of Science and Technology, Meiji University Kawasaki, Japan
| | - Makoto Asashima
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Yasushi Hojo
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan ; Bioinformatics Project (BIRD), Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Core Research for Evolutional Science and Technology Project of Japan Science and Technology Agency, The University of Tokyo Meguro, Japan
| | - Muneki Ikeda
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Yoshimasa Komatsuzaki
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Yuuki Ooishi
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Suguru Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan ; Bioinformatics Project (BIRD), Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Core Research for Evolutional Science and Technology Project of Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; National MEXT Project in Special Coordinate Funds for Promoting Science and Technology, The University of Tokyo Meguro, Japan
| |
Collapse
|
26
|
Gancarz-Kausch AM, Schroeder GL, Panganiban C, Adank D, Humby MS, Kausch MA, Clark SD, Dietz DM. Transforming growth factor beta receptor 1 is increased following abstinence from cocaine self-administration, but not cocaine sensitization. PLoS One 2013; 8:e83834. [PMID: 24386286 PMCID: PMC3875479 DOI: 10.1371/journal.pone.0083834] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/15/2013] [Indexed: 12/24/2022] Open
Abstract
The addicted phenotype is characterized as a long-lasting, chronically relapsing disorder that persists following long periods of abstinence, suggesting that the underlying molecular changes are stable and endure for long periods even in the absence of drug. Here, we investigated Transforming Growth Factor-Beta Type I receptor (TGF-β R1) expression in the nucleus accumbens (NAc) following periods of withdrawal from cocaine self-administration (SA) and a sensitizing regimen of non-contingent cocaine. Rats were exposed to either (i) repeated systemic injections (cocaine or saline), or (ii) self-administration (cocaine or saline) and underwent a period of forced abstinence (either 1 or 7 days of drug cessation). Withdrawal from cocaine self-administration resulted in an increase in TGF-β R1 protein expression in the NAc compared to saline controls. This increase was specific for volitional cocaine intake as no change in expression was observed following a sensitizing regimen of experimenter-administered cocaine. These findings implicate TGF-β signaling as a novel potential therapeutic target for treating drug addiction.
Collapse
Affiliation(s)
- Amy M. Gancarz-Kausch
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
- Research Institute on Addictions, University at Buffalo, Buffalo, New York, United States of America
| | - Gabrielle L. Schroeder
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
| | - Clarisse Panganiban
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
| | - Danielle Adank
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
| | - Monica S. Humby
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
| | - Michael A. Kausch
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
| | - Stewart D. Clark
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
- Research Institute on Addictions, University at Buffalo, Buffalo, New York, United States of America
| | - David M. Dietz
- Department of Pharmacology & Toxicology, University at Buffalo, Buffalo, New York, United States of America
- Research Institute on Addictions, University at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
27
|
Tapia-González S, Muñoz MD, Cuartero MI, Sánchez-Capelo A. Smad3 is required for the survival of proliferative intermediate progenitor cells in the dentate gyrus of adult mice. Cell Commun Signal 2013; 11:93. [PMID: 24330661 PMCID: PMC4029396 DOI: 10.1186/1478-811x-11-93] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/06/2013] [Indexed: 12/16/2022] Open
Abstract
Background New neurons are continuously being generated in the adult hippocampus, a phenomenon that is regulated by external stimuli, such as learning, memory, exercise, environment or stress. However, the molecular mechanisms underlying neuron production and how they are integrated into existing circuits under such physiological conditions remain unclear. Indeed, the intracellular modulators that transduce the extracellular signals are not yet fully understood. Results We show that Smad3, an intracellular molecule involved in the transforming growth factor (TGF)-β signaling cascade, is strongly expressed by granule cells in the dentate gyrus (DG) of adult mice, although the loss of Smad3 in null mutant mice does not affect their survival. Smad3 is also expressed by adult progenitor cells in the subgranular zone (SGZ) and more specifically, it is first expressed by Type 2 cells (intermediate progenitor cells). Its expression persists through the distinct cell stages towards that of the mature neuron. Interestingly, proliferative intermediate progenitor cells die in Smad3 deficiency, which is associated with a large decrease in the production of newborn neurons in Smad3 deficient mice. Smad3 signaling appears to influence adult neurogenesis fulfilling distinct roles in the rostral and mid-caudal regions of the DG. In rostral areas, Smad3 deficiency increases proliferation and promotes the cell cycle exit of undifferentiated progenitor cells. By contrast, Smad3 deficiency impairs the survival of newborn neurons in the mid-caudal region of the DG at early proliferative stages, activating apoptosis of intermediate progenitor cells. Furthermore, long-term potentiation (LTP) after high frequency stimulation (HFS) to the medial perforant path (MPP) was abolished in the DG of Smad3-deficient mice. Conclusions These data show that endogenous Smad3 signaling is central to neurogenesis and LTP induction in the adult DG, these being two forms of hippocampal brain plasticity related to learning and memory that decline with aging and as a result of neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Amelia Sánchez-Capelo
- CIBERNED - Ser, Neurobiología-Investigación, Hospital Universitario Ramón y Cajal-IRYCIS, Ctra, Colmenar Viejo Km 9, 28034 Madrid, Spain.
| |
Collapse
|
28
|
Gu W, Fukuda T, Isaji T, Hashimoto H, Wang Y, Gu J. α1,6-Fucosylation regulates neurite formation via the activin/phospho-Smad2 pathway in PC12 cells: the implicated dual effects of Fut8 for TGF-β/activin-mediated signaling. FASEB J 2013; 27:3947-58. [PMID: 23796784 DOI: 10.1096/fj.12-225805] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It is well known that α1,6-fucosyltransferase (Fut8) and its products, α1,6-fucosylated N-glycans, are highly expressed in brain tissue. Recently, we reported that Fut8-knockout mice exhibited multiple behavioral abnormalities with a schizophrenia-like phenotype, suggesting that α1,6-fucosylation plays important roles in the brain and neuron system. In the present study, we screened several neural cell lines and found that PC12 cells express the highest levels of α1,6-fucosylation. The knockdown (KD) of Fut8 promoted a significant enhancement of neurite formation and induction of neurofilament expression. Surprisingly, the levels of phospho-Smad2 were greatly increased in the KD cells. Finally, we found that the activin-mediated signal pathway was essential for these changes in KD cells. Exogenous activin, not TGF-β1, induced neurite outgrowth and phospho-Smad2. In addition, the α1,6-fucosylation level on the activin receptors was greatly decreased in KD cells, while the total expression level was unchanged, suggesting that α1,6-fucosylation negatively regulated activin-mediated signaling. Furthermore, inhibition of activin receptor-mediated signaling or restoration of Fut8 expression rescued cell morphology and phospho-Smad2 levels, which were enhanced in KD cells. Considering the fact that α1,6-fucosylation is important for TGF-β-mediated signaling, the results of this study strongly suggest that Fut8 plays a dual role in TGF-β/activin-mediated signaling.
Collapse
Affiliation(s)
- Wei Gu
- 1Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai Miyagi, 981-8558, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Ganea K, Menke A, Schmidt MV, Lucae S, Rammes G, Liebl C, Harbich D, Sterlemann V, Storch C, Uhr M, Holsboer F, Binder EB, Sillaber I, Müller MB. Convergent animal and human evidence suggests the activin/inhibin pathway to be involved in antidepressant response. Transl Psychiatry 2012; 2:e177. [PMID: 23092981 PMCID: PMC3565812 DOI: 10.1038/tp.2012.104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Despite the overt need for improved treatment modalities in depression, efforts to develop conceptually novel antidepressants have been relatively unsuccessful so far. Here we present a translational approach combining results from hypothesis-free animal experiments with data from a genetic association study in depression. Comparing genes regulated by chronic paroxetine treatment in the mouse hippocampus with genes showing nominally significant association with antidepressant treatment response in two pharmacogenetic studies, the activin pathway was the only one to show this dual pattern of association and therefore selected as a candidate. We examined the regulation of activin A and activin receptor type IA mRNA following antidepressant treatment. We investigated the effects of stereotaxic infusion of activin into the hippocampus and the amygdala in a behavioural model of depression. To analyse whether variants in genes in the activin signalling pathway predict antidepressant treatment response, we performed a human genetic association study. Significant changes in the expression of genes in the activin signalling pathway were observed following 1 and 4 weeks of treatment. Injection of activin A into the hippocampus exerts acute antidepressant-like effects. Polymorphisms in the betaglycan gene, a co-receptor mediating functional antagonism of activin signalling, significantly predict treatment outcome in our system-wide pharmacogenetics study in depression. We provide convergent evidence from mouse and human data that genes in the activin signalling pathway are promising novel candidates involved in the neurobiogical mechanisms underlying antidepressant mechanisms of action. Further, our data suggest this pathway to be a target for more rapid-acting antidepressants in the future.
Collapse
Affiliation(s)
- K Ganea
- Max Planck Institute of Psychiatry, Munich, Germany
| | - A Menke
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M V Schmidt
- Max Planck Institute of Psychiatry, Munich, Germany
| | - S Lucae
- Max Planck Institute of Psychiatry, Munich, Germany
| | - G Rammes
- Max Planck Institute of Psychiatry, Munich, Germany,Department of Anesthesiology, Technische Universtität, Munich, Germany
| | - C Liebl
- Max Planck Institute of Psychiatry, Munich, Germany
| | - D Harbich
- Max Planck Institute of Psychiatry, Munich, Germany
| | - V Sterlemann
- Max Planck Institute of Psychiatry, Munich, Germany
| | - C Storch
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M Uhr
- Max Planck Institute of Psychiatry, Munich, Germany
| | - F Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany
| | - E B Binder
- Max Planck Institute of Psychiatry, Munich, Germany
| | - I Sillaber
- Phenoquest AG, Martinsried/Munich, Munich, Germany
| | - M B Müller
- Max Planck Institute of Psychiatry, Munich, Germany,Molecular Stress Physiology, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany. E-mail:
| |
Collapse
|
30
|
Low gene expression of bone morphogenetic protein 7 in brainstem astrocytes in major depression. Int J Neuropsychopharmacol 2012; 15:855-68. [PMID: 21896235 DOI: 10.1017/s1461145711001350] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The noradrenergic locus coeruleus (LC) is the principal source of brain norepinephrine, a neurotransmitter thought to play a major role in the pathology of major depressive disorder (MDD) and in the therapeutic action of many antidepressant drugs. The goal of this study was to identify potential mediators of brain noradrenergic dysfunction in MDD. Bone morphogenetic protein 7 (BMP7), a member of the transforming growth factor-β superfamily, is a critical mediator of noradrenergic neuron differentiation during development and has neurotrophic and neuroprotective effects on mature catecholaminergic neurons. Real-time PCR of reversed transcribed RNA isolated from homogenates of LC tissue from 12 matched pairs of MDD subjects and psychiatrically normal control subjects revealed low levels of BMP7 gene expression in MDD. No differences in gene expression levels of other members of the BMP family were observed in the LC, and BMP7 gene expression was normal in the prefrontal cortex and amygdala in MDD subjects. Laser capture microdissection of noradrenergic neurons, astrocytes, and oligodendrocytes from the LC revealed that BMP7 gene expression was highest in LC astrocytes relative to the other cell types, and that the MDD-associated reduction in BMP7 gene expression was limited to astrocytes. Rats exposed to chronic social defeat exhibited a similar reduction in BMP7 gene expression in the LC. BMP7 has unique developmental and trophic actions on catecholamine neurons and these findings suggest that reduced astrocyte support for pontine LC neurons may contribute to pathology of brain noradrenergic neurons in MDD.
Collapse
|
31
|
Baj G, D'Alessandro V, Musazzi L, Mallei A, Sartori CR, Sciancalepore M, Tardito D, Langone F, Popoli M, Tongiorgi E. Physical exercise and antidepressants enhance BDNF targeting in hippocampal CA3 dendrites: further evidence of a spatial code for BDNF splice variants. Neuropsychopharmacology 2012; 37:1600-11. [PMID: 22318196 PMCID: PMC3358751 DOI: 10.1038/npp.2012.5] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is encoded by multiple BDNF transcripts, whose function is unclear. We recently showed that a subset of BDNF transcripts can traffic into distal dendrites in response to electrical activity, while others are segregated into the somatoproximal domains. Physical exercise and antidepressant treatments exert their beneficial effects through upregulation of BDNF, which is required to support survival and differentiation of newborn dentate gyrus (DG) neurons. While these DG processes are required for the antidepressant effect, a role for CA1 in antidepressant action has been excluded, and the effect on CA3 neurons remains unclear. Here, we show for the first time that physical exercise and antidepressants induce local increase of BDNF in CA3. Voluntary physical exercise for 28 consecutive days, or 2-week treatment with 10 mg/kg per day fluoxetine or reboxetine, produced a global increase of BDNF mRNA and protein in the neuronal somata of the whole hippocampus and a specific increase of BDNF in dendrites of CA3 neurons. This increase was accounted for by BDNF exon 6 variant. In cultured hippocampal neurons, application of serotonin or norepinephrine (10-50 μM) induced increase in synaptic transmission and targeting of BDNF mRNA in dendrites. The increased expression of BDNF in CA3 dendrites following antidepressants or exercise further supports the neurotrophin hypothesis of antidepressants action and confirms that the differential subcellular localization of BDNF mRNA splice variants provides a spatial code for a selective expression of BDNF in specific subcellular districts. This selective expression may be exploited to design more specific antidepressants.
Collapse
Affiliation(s)
- Gabriele Baj
- Department of Life Sciences, BRAIN Centre for Neuroscience, University of Trieste, Trieste, Italy
| | - Valentina D'Alessandro
- Department of Life Sciences, BRAIN Centre for Neuroscience, University of Trieste, Trieste, Italy
| | - Laura Musazzi
- Center of Neuropharmacology—Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Alessandra Mallei
- Center of Neuropharmacology—Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Cesar R Sartori
- Department of Physiology and Biophysics, Institute of Biology, State University of Campinas, UNICAMP, Campinas, Brazil
| | - Marina Sciancalepore
- Department of Life Sciences, BRAIN Centre for Neuroscience, University of Trieste, Trieste, Italy
| | - Daniela Tardito
- Center of Neuropharmacology—Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Francesco Langone
- Department of Physiology and Biophysics, Institute of Biology, State University of Campinas, UNICAMP, Campinas, Brazil
| | - Maurizio Popoli
- Center of Neuropharmacology—Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Enrico Tongiorgi
- Department of Life Sciences, BRAIN Centre for Neuroscience, University of Trieste, Trieste, Italy,Department of Life Sciences, BRAIN Centre for Neuroscience, University of Trieste, Via Giorgieri, 5, Building Q, Trieste 34127, Italy, Tel: +39 040 558 8724, Fax: +39 040 558 2425, E-mail:
| |
Collapse
|
32
|
Miller MC, Lambert-Messerlian GM, Eklund EE, Heath NL, Donahue JE, Stopa EG. Expression of inhibin/activin proteins and receptors in the human hypothalamus and basal forebrain. J Neuroendocrinol 2012; 24:962-72. [PMID: 22296042 DOI: 10.1111/j.1365-2826.2012.02289.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The inhibin/activin family of proteins is known to have a broad distribution of synthesis and expression in many species, as well as a variety of functions in reproductive and other physiological systems. Yet, our knowledge regarding the production and function of inhibin and activin in the central nervous system is relatively limited, especially in humans. The present study aimed to explore the distribution of inhibin/activin protein subunits and receptors in the adult human brain. The human hypothalamus and surrounding basal forebrain was examined using post-mortem tissues from 29 adults. Immunocytochemical studies were conducted with antibodies directed against the inhibin/activin α, βA, and βB subunits, betaglycan and the activin type IIA and IIB receptors. An immunoassay was also utilised to measure dimeric inhibin A and B levels in tissue homogenates of the infundibulum of the hypothalamus. Robust βA subunit immunoreactivity was present in the paraventricular, supraoptic, lateral hypothalamic, infundibular, dorsomedial and suprachiasmatic nuclei of the hypothalamus, in the basal ganglia, and in the nucleus basalis of Meynert. A similar staining distribution was noted for the βB subunit, betaglycan and the type II receptor antibodies, whereas α subunit staining was not detected in any of the major anatomical regions of the human brain. Inhibin B immunoreactivity was present in all tissues, whereas inhibin A levels were below detectable limits. These studies show for the first time that the inhibin/activin protein subunits and receptors can be co-localised in the human brain, implicating potential, diverse neural functions.
Collapse
Affiliation(s)
- M C Miller
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | | | | | | | | |
Collapse
|
33
|
Satvat E, Gheidi A, Voll S, Odintsova IV, Marrone DF. Location is everything: neurons born during fluoxetine treatment accumulate in regions that do not support spatial learning. Neuropharmacology 2011; 62:1627-33. [PMID: 22182782 DOI: 10.1016/j.neuropharm.2011.11.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 11/28/2011] [Accepted: 11/30/2011] [Indexed: 12/30/2022]
Abstract
It is well known that antidepressants both improve mood and increase the rate at which the dentate gyrus (DG) generates new neurons. In addition to the implications of neurogenesis for mood regulation, the production and survival of granule cells has also been implicated in learning and memory. Despite this evidence, the results of studies on the effect of antidepressants on memory have been mixed. A critical piece of data that may be missing from previous studies, however, is insight into (a) the location that newborn neurons migrate to following fluoxetine administration and (b) their ability to express normal patterns of activity-related genes. Here we demonstrate a finding that may resolve the discrepancy in the effects fluoxetine-induced neurogenesis on mood and memory: after 5 weeks delay, the net additional neurons generated in animals given the antidepressant fluoxetine during treatment are functionally normal, but preferentially accumulate (due to changes in migration and/or survival) in an area of the DG that is not recruited by spatial memory tasks.
Collapse
Affiliation(s)
- Elham Satvat
- Dept. of Psychology, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | | | | | | | | |
Collapse
|
34
|
Krieglstein K, Zheng F, Unsicker K, Alzheimer C. More than being protective: functional roles for TGF-β/activin signaling pathways at central synapses. Trends Neurosci 2011; 34:421-9. [PMID: 21742388 DOI: 10.1016/j.tins.2011.06.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/30/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022]
Abstract
It is becoming increasingly clear that members of the transforming growth factor-β (TGF-β) family have roles in the central nervous system that extend beyond their well-established roles as neurotrophic and neuroprotective factors. Recent findings have indicated that the TGF-β signaling pathways are involved in the modulation of both excitatory and inhibitory synaptic transmission in the adult mammalian brain. In this review, we discuss how TGF-β, bone morphogenetic protein and activin signaling at central synapses modulate synaptic plasticity, cognition and affective behavior. We also discuss the implications of these findings for the molecular understanding and potential treatment of neuropsychiatric diseases, such as anxiety, depression and other neurological disorders.
Collapse
Affiliation(s)
- Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | | | | | | |
Collapse
|
35
|
Depino AM, Lucchina L, Pitossi F. Early and adult hippocampal TGF-β1 overexpression have opposite effects on behavior. Brain Behav Immun 2011; 25:1582-91. [PMID: 21640817 DOI: 10.1016/j.bbi.2011.05.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 04/29/2011] [Accepted: 05/16/2011] [Indexed: 10/18/2022] Open
Abstract
TGF-β1 is an anti-inflammatory cytokine that is augmented in the brain of autistic patients and that can affect brain development. In this work, we studied the effects of overexpressing TGF-β1 in the dentate gyrus of adult or young mice on behavior. TGF-β1 overexpression during postnatal development led to a long-term decrease in social interaction and to long-term increases in self-grooming and depression-related behaviors. Our analysis shows that these behavioral changes correlate with the long-term downregulation of TGF-β1 and IL-6 expression in the dentate gyrus, as well as to decreases in the mRNA levels of the synaptic protein neuroligin 3 and in the number of Reelin-positive neurons in the dentate gyrus. In contrast, chronic expression of TGF-β1 during adulthood led to transient opposite effects on these behaviors. These results show a central role of hippocampal TGF-β1 in the programming and modulation of social interaction, repetitive behavior and depression-related behavior. Finally, our data suggest a role of hippocampal TGF-β1 and early-life neuroinflammation in the development of the behavioral alterations observed in autism spectrum disorders.
Collapse
Affiliation(s)
- Amaicha Mara Depino
- Institute for Physiology, Molecular Biology and Neurosciences, CONICET-UBA, C1428EHA Buenos Aires, Argentina.
| | | | | |
Collapse
|
36
|
Chronic antidepressant treatments induce a time-dependent up-regulation of AMPA receptor subunit protein levels. Neurochem Int 2011; 59:896-905. [DOI: 10.1016/j.neuint.2011.07.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 06/30/2011] [Accepted: 07/28/2011] [Indexed: 12/15/2022]
|
37
|
Barbon A, Orlandi C, La Via L, Caracciolo L, Tardito D, Musazzi L, Mallei A, Gennarelli M, Racagni G, Popoli M, Barlati S. Antidepressant treatments change 5-HT2C receptor mRNA expression in rat prefrontal/frontal cortex and hippocampus. Neuropsychobiology 2011; 63:160-8. [PMID: 21228608 DOI: 10.1159/000321593] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 09/27/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Compelling evidence would suggest the involvement of the serotonin 2C receptor in the pathophysiology of affective disorders and in the action of antidepressants. We analyzed the time course of 5-HT2C receptor (5-HTR2C) mRNA expression during antidepressant treatment in the prefrontal/frontal cortex (P/FC) and in the hippocampus (HC) of rats chronically treated with fluoxetine (a selective serotonin reuptake inhibitor) and reboxetine (a selective noradrenaline reuptake inhibitor). We also analyzed the 5-HTR2C RNA-editing levels at the sites called A, B, C, C' and D, which are known to modulate 5-HTR2C receptor function. RESULTS The expression profile of 5-HTR2C mRNA was modified during treatment with both antidepressants. In particular, we found a general down-regulation of 5-HTR2C mRNA expression in P/FC, which became significant after 3 weeks of treatment with both antidepressants and persisted after a fourth week of drug withdrawal (-46% with fluoxetine, -41% with reboxetine, p < 0.05). In HC, however, reboxetine induced significant down-regulation (-56%, p < 0.05) of 5-HTR2C mRNA after 3 weeks, while fluoxetine induced threefold up-regulation (p < 0.01) by the 2nd and 3rd week, returning to the base level after drug withdrawal of both antidepressants. Moreover, the frequency of 5-HTR2C-edited isoforms showed no significant alterations, although analysis of the RNA-editing level at the single editing sites showed small decreases in the C' and D sites induced by reboxetine in P/FC. CONCLUSION Our results suggest that chronic administration of antidepressants in rats slightly modifies the editing levels of 5-HT2C receptor but has considerable influence on its mRNA expression patterns in a way that is area- and time-specific.
Collapse
Affiliation(s)
- Alessandro Barbon
- Division of Biology and Genetics, Department of Biomedical Sciences and Biotechnologies and National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Padilla E, Shumake J, Barrett DW, Sheridan EC, Gonzalez-Lima F. Mesolimbic effects of the antidepressant fluoxetine in Holtzman rats, a genetic strain with increased vulnerability to stress. Brain Res 2011; 1387:71-84. [PMID: 21376019 DOI: 10.1016/j.brainres.2011.02.080] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 02/24/2011] [Accepted: 02/24/2011] [Indexed: 11/26/2022]
Abstract
This is the first metabolic mapping study of the effects of fluoxetine after learned helplessness training. Antidepressants are the most commonly prescribed medications, but the regions underlying treatment effects in affectively disordered brains are poorly understood. We hypothesized the antidepressant action of fluoxetine would produce adaptations in mesolimbic regions after 2 weeks of treatment. We used Holtzman rats, a genetic strain showing susceptibility to novelty-evoked hyperactivity and stress-evoked helplessness, to map regional brain metabolic effects caused by fluoxetine treatment. Animals underwent learned helplessness, and subsequently immobility time was scored in the forced swim test (FST). On the next day, animals began receiving 2 weeks of fluoxetine (5mg/kg/day) or vehicle and were retested in the FST at the end of drug treatment. Antidepressant behavioral effects of fluoxetine were analyzed using a ratio of immobility during pre- and post-treatment FST sessions. Brains were analyzed for regional metabolic activity using quantitative cytochrome oxidase histochemistry as in our previous study using congenitally helpless rats. Fluoxetine exerted a protective effect against FST-induced immobility behavior in Holtzman rats. Fluoxetine also caused a significant reduction in the mean regional metabolism of the nucleus accumbens shell and the ventral hippocampus as compared to vehicle-treated subjects. Additional networks affected by fluoxetine treatment included the prefrontal-cingulate cortex and brainstem nuclei linked to depression (e.g., habenula, dorsal raphe and interpeduncular nucleus). We concluded that corticolimbic regions such as the prefrontal-cingulate cortex, nucleus accumbens, ventral hippocampus and key brainstem nuclei represent important contributors to the neural network mediating fluoxetine antidepressant action.
Collapse
Affiliation(s)
- Eimeira Padilla
- Department of Psychology, University of Texas at Austin, 1 University Station A8000, Austin, TX 78712, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Activins, which are members of the TGF-β superfamily, were initially isolated from gonads and served as modulators of follicle-stimulating hormone secretion. Activins regulate various biological functions, including induction of the dorsal mesoderm, craniofacial development, and differentiation of numerous cell types. Activin receptors are highly expressed in neuronal cells, and activin mRNA expression is upregulated by neuronal activity. Activins also exhibit neuroprotective action during excitotoxic brain injury. However, very little is known about the functional roles of activins in the brain. We recently generated various types of transgenic mice, demonstrating that activins regulate spine formation, behavioral activity, anxiety, adult neurogenesis, late-phase long-term potentiation, and maintenance of long-term memory. The present chapter describes recent progress in the study of the role of activin in the brain.
Collapse
Affiliation(s)
- Hiroshi Ageta
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Aichi, Japan
| | | |
Collapse
|
40
|
Gajos-Michniewicz A, Piastowska AW, Russell JA, Ochedalski T. Follistatin as a potent regulator of bone metabolism. Biomarkers 2010; 15:563-74. [PMID: 20569048 DOI: 10.3109/1354750x.2010.495786] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Follistatin is a monomeric glycoprotein, distributed in a wide range of tissues. Recent work has demonstrated that this protein is a pluripotential molecule that has no structural similarity but is functionally associated with members of the transforming growth factor (TGF)-β superfamily, which indicates its wide range of action. Members of the TGF-β superfamily, especially activins and bone morphogenetic proteins are involved in bone metabolism. They play an important role in bone physiology, influencing bone growth, turnover, bone formation and cartilage induction. As follistatin is considered to be the antagonist of the TGF-β superfamily members, it plays an important role in bone metabolism and development.
Collapse
Affiliation(s)
- A Gajos-Michniewicz
- Department of Comparative Endocrinology, Medical University of Lodz, Poland.
| | | | | | | |
Collapse
|
41
|
Canonical TGF-beta signaling is required for the balance of excitatory/inhibitory transmission within the hippocampus and prepulse inhibition of acoustic startle. J Neurosci 2010; 30:6025-35. [PMID: 20427661 DOI: 10.1523/jneurosci.0789-10.2010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Smad4 is a unique nuclear transducer for all TGF-beta signaling pathways and regulates gene transcription during development and tissue homeostasis. To elucidate the postnatal role of TGF-beta signaling in the mammalian brain, we generated forebrain-specific Smad4 knock-out mice. Surprisingly, the mutants showed no alteration in long-term potentiation and water maze, suggesting that Smad4 is not required for spatial learning and memory. However, these mutant mice did show enhancement of paired-pulse facilitation in excitatory synaptic transmission and stronger paired-pulse depression of GABA(A) currents in the hippocampus. The alteration of hippocampal electrophysiology correlated with mouse hyperactivity in homecage and open field tests. Mutant mice also showed overgrooming as well as deficits of prepulse inhibition, a widely used endophenotype of schizophrenia. With a specific real-time PCR array focused on TGF-beta signaling pathway, we identified a novel regulation mechanism of the pathway in the hippocampal neurons, in which Smad4-mediated signaling suppresses the level of extracellular antagonism of TGF-beta ligands through transcriptional regulation of follistatin, a selective inhibitor to activin/TGF-beta signaling in the hippocampus. In summary, we suggest that the canonical TGF-beta signaling pathway is critical for use-dependent modulation of GABA(A) synaptic transmission and dendritic homeostasis; furthermore, a disruption in the balance of the excitatory and inhibitory hippocampal network can result in psychiatric-like behavior.
Collapse
|
42
|
Suzuki K, Kobayashi T, Funatsu O, Morita A, Ikekita M. Activin A induces neuronal differentiation and survival via ALK4 in a SMAD-independent manner in a subpopulation of human neuroblastomas. Biochem Biophys Res Commun 2010; 394:639-45. [DOI: 10.1016/j.bbrc.2010.03.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 03/06/2010] [Indexed: 10/19/2022]
|
43
|
Ageta H, Ikegami S, Miura M, Masuda M, Migishima R, Hino T, Takashima N, Murayama A, Sugino H, Setou M, Kida S, Yokoyama M, Hasegawa Y, Tsuchida K, Aosaki T, Inokuchi K. Activin plays a key role in the maintenance of long-term memory and late-LTP. Learn Mem 2010; 17:176-85. [PMID: 20332189 DOI: 10.1101/lm.16659010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A recent study has revealed that fear memory may be vulnerable following retrieval, and is then reconsolidated in a protein synthesis-dependent manner. However, little is known about the molecular mechanisms of these processes. Activin betaA, a member of the TGF-beta superfamily, is increased in activated neuronal circuits and regulates dendritic spine morphology. To clarify the role of activin in the synaptic plasticity of the adult brain, we examined the effect of inhibiting or enhancing activin function on hippocampal long-term potentiation (LTP). We found that follistatin, a specific inhibitor of activin, blocked the maintenance of late LTP (L-LTP) in the hippocampus. In contrast, administration of activin facilitated the maintenance of early LTP (E-LTP). We generated forebrain-specific activin- or follistatin-transgenic mice in which transgene expression is under the control of the Tet-OFF system. Maintenance of hippocampal L-LTP was blocked in the follistatin-transgenic mice. In the contextual fear-conditioning test, we found that follistatin blocked the formation of long-term memory (LTM) without affecting short-term memory (STM). Furthermore, consolidated memory was selectively weakened by the expression of follistatin during retrieval, but not during the maintenance phase. On the other hand, the maintenance of memory was also influenced by activin overexpression during the retrieval phase. Thus, the level of activin in the brain during the retrieval phase plays a key role in the maintenance of long-term memory.
Collapse
Affiliation(s)
- Hiroshi Ageta
- Mitsubishi Kagaku Institute of Life Sciences, MITILS, Machida, Tokyo 194-8511, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kinsler R, Taylor MM, Flores NM, Leffert JJ, Beech RD. Altered response to antidepressant treatment in FoxG1 heterozygous knockout mice. Synapse 2010; 64:169-71. [PMID: 19852072 DOI: 10.1002/syn.20737] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Evidence from a variety of sources suggests that structural alterations in the brain, including neurogenesis, may play a role in both the pathogenesis of mood disorders and the mechanism of action of antidepressants. Previous studies have implicated both the transforming growth factor-beta (TGF-beta), and the phosphatidyl inositol-3 kinase (PI3K)-Akt pathways in the neurogenesis-promoting and behavioral properties of antidepressants. Forkhead box protein G1 (FoxG1) is a major regulator of both of these pathways, and FoxG1 heterozygous null mice (FoxG1+/-) have previously been reported to have deficits in adult hippocampal neurogenesis and behavioral abnormalities including deficits in contextual fear learning. However the role of FoxG1, if any, in the response to antidepressants has not been previously investigated.To investigate the role of the FoxG1 gene in the behavioral and neurogenic properties of antidepressants, we tested FoxG1+/- mice and littermate controls in two different rodent models of antidepressant action: the tail suspension test and the forced swim test. FoxG1+/- mice showed no response to antidepressants in either of these tests. These results suggest that normal levels of FoxG1 may be required for the behavioral response to antidepressants.
Collapse
Affiliation(s)
- Rebecca Kinsler
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | | | | | | |
Collapse
|
45
|
Musazzi L, Milanese M, Farisello P, Zappettini S, Tardito D, Barbiero VS, Bonifacino T, Mallei A, Baldelli P, Racagni G, Raiteri M, Benfenati F, Bonanno G, Popoli M. Acute stress increases depolarization-evoked glutamate release in the rat prefrontal/frontal cortex: the dampening action of antidepressants. PLoS One 2010; 5:e8566. [PMID: 20052403 PMCID: PMC2797327 DOI: 10.1371/journal.pone.0008566] [Citation(s) in RCA: 190] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 12/06/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Behavioral stress is recognized as a main risk factor for neuropsychiatric diseases. Converging evidence suggested that acute stress is associated with increase of excitatory transmission in certain forebrain areas. Aim of this work was to investigate the mechanism whereby acute stress increases glutamate release, and if therapeutic drugs prevent the effect of stress on glutamate release. METHODOLOGY/FINDINGS Rats were chronically treated with vehicle or drugs employed for therapy of mood/anxiety disorders (fluoxetine, desipramine, venlafaxine, agomelatine) and then subjected to unpredictable footshock stress. Acute stress induced marked increase in depolarization-evoked release of glutamate from synaptosomes of prefrontal/frontal cortex in superfusion, and the chronic drug treatments prevented the increase of glutamate release. Stress induced rapid increase in the circulating levels of corticosterone in all rats (both vehicle- and drug-treated), and glutamate release increase was blocked by previous administration of selective antagonist of glucocorticoid receptor (RU 486). On the molecular level, stress induced accumulation of presynaptic SNARE complexes in synaptic membranes (both in vehicle- and drug-treated rats). Patch-clamp recordings of pyramidal neurons in the prefrontal cortex revealed that stress increased glutamatergic transmission through both pre- and postsynaptic mechanisms, and that antidepressants may normalize it by reducing release probability. CONCLUSIONS/SIGNIFICANCE Acute footshock stress up-regulated depolarization-evoked release of glutamate from synaptosomes of prefrontal/frontal cortex. Stress-induced increase of glutamate release was dependent on stimulation of glucocorticoid receptor by corticosterone. Because all drugs employed did not block either elevation of corticosterone or accumulation of SNARE complexes, the dampening action of the drugs on glutamate release must be downstream of these processes. This novel effect of antidepressants on the response to stress, shown here for the first time, could be related to the therapeutic action of these drugs.
Collapse
Affiliation(s)
- Laura Musazzi
- Department of Pharmacological Sciences, Center of Neuropharmacology and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Marco Milanese
- Department of Experimental Medicine, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Center of Excellence for Biomedical Research and National Institute of Neuroscience, Genova, Italy
| | - Pasqualina Farisello
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova and National Institute of Neuroscience, Genova, Italy
| | - Simona Zappettini
- Department of Experimental Medicine, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Center of Excellence for Biomedical Research and National Institute of Neuroscience, Genova, Italy
| | - Daniela Tardito
- Department of Pharmacological Sciences, Center of Neuropharmacology and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Valentina S. Barbiero
- Department of Pharmacological Sciences, Center of Neuropharmacology and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Tiziana Bonifacino
- Department of Experimental Medicine, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Center of Excellence for Biomedical Research and National Institute of Neuroscience, Genova, Italy
| | - Alessandra Mallei
- Department of Pharmacological Sciences, Center of Neuropharmacology and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Pietro Baldelli
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova and National Institute of Neuroscience, Genova, Italy
| | - Giorgio Racagni
- Department of Pharmacological Sciences, Center of Neuropharmacology and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
- Department of Experimental Medicine, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico San Giovanni di Dio - Fatebenefratelli, Brescia, Italy
- Center of Excellence for Biomedical Research and National Institute of Neuroscience, Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova and National Institute of Neuroscience, Genova, Italy
| | - Maurizio Raiteri
- Department of Experimental Medicine, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Center of Excellence for Biomedical Research and National Institute of Neuroscience, Genova, Italy
| | - Fabio Benfenati
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova and National Institute of Neuroscience, Genova, Italy
| | - Giambattista Bonanno
- Department of Experimental Medicine, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Center of Excellence for Biomedical Research and National Institute of Neuroscience, Genova, Italy
| | - Maurizio Popoli
- Department of Pharmacological Sciences, Center of Neuropharmacology and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| |
Collapse
|
46
|
Early induction of CREB activation and CREB-regulating signalling by antidepressants. Int J Neuropsychopharmacol 2009; 12:1367-81. [PMID: 19400982 DOI: 10.1017/s1461145709000376] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Converging evidence points to adaptive changes in neuroplasticity and gene expression as mediators of therapeutic action of antidepressants. Activation of cAMP response-element binding protein (CREB) and CREB-regulating signalling are considered main effectors in these mechanisms. We analysed the temporal profile of intracellular changes induced by antidepressants, by measuring activation of major CREB-regulating signalling cascades and activation (Ser133 phosphorylation) of CREB. The main aims of the study were to investigate how these different variables are modulated with time, whether stronger activation of signalling cascades corresponds to stronger activation of CREB, and whether these changes are different in distinct brain areas. Rat groups were treated for 1, 2 or 3 wk with the antidepressants fluoxetine or reboxetine; in additional groups drug treatment was followed by a washout week (3+1). Activation of CREB and major effectors in signalling cascades were analysed by Western blot analysis with phospho-antibodies, in nuclear and cytosolic fractions from hippocampus and prefrontal/frontal cortex (P/FC). Surprisingly, CREB activation was already maximal after 1-wk treatment. In hippocampus early and stronger CREB activation was consistent with early and stronger activation of signalling. For both drugs, the profile of activation in P/FC was different from that observed in hippocampus. The results also showed that, contrary to the activatory role of MAP-ERKs and CaM kinase IV, nuclear alphaCaM kinase II was inactivated in parallel with activation of CREB.
Collapse
|
47
|
Nishino Y, Ooishi R, Kurokawa S, Fujino K, Murakami M, Madarame H, Hashimoto O, Sugiyama K, Funaba M. Gene expression of the TGF-β family in rat brain infected with Borna disease virus. Microbes Infect 2009; 11:737-43. [DOI: 10.1016/j.micinf.2009.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/02/2009] [Accepted: 04/07/2009] [Indexed: 11/17/2022]
|
48
|
Tsuchida K, Nakatani M, Hitachi K, Uezumi A, Sunada Y, Ageta H, Inokuchi K. Activin signaling as an emerging target for therapeutic interventions. Cell Commun Signal 2009; 7:15. [PMID: 19538713 PMCID: PMC2713245 DOI: 10.1186/1478-811x-7-15] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 06/18/2009] [Indexed: 01/24/2023] Open
Abstract
After the initial discovery of activins as important regulators of reproduction, novel and diverse roles have been unraveled for them. Activins are expressed in various tissues and have a broad range of activities including the regulation of gonadal function, hormonal homeostasis, growth and differentiation of musculoskeletal tissues, regulation of growth and metastasis of cancer cells, proliferation and differentiation of embryonic stem cells, and even higher brain functions. Activins signal through a combination of type I and II transmembrane serine/threonine kinase receptors. Activin receptors are shared by multiple transforming growth factor-β (TGF-β) ligands such as myostatin, growth and differentiation factor-11 and nodal. Thus, although the activity of each ligand is distinct, they are also redundant, both physiologically and pathologically in vivo. Activin receptors activated by ligands phosphorylate the receptor-regulated Smads for TGF-β, Smad2 and 3. The Smad proteins then undergo multimerization with the co-mediator Smad4, and translocate into the nucleus to regulate the transcription of target genes in cooperation with nuclear cofactors. Signaling through receptors and Smads is controlled by multiple mechanisms including phosphorylation and other posttranslational modifications such as sumoylation, which affect potein localization, stability and transcriptional activity. Non-Smad signaling also plays an important role in activin signaling. Extracellularly, follistatin and related proteins bind to activins and related TGF-β ligands, and control the signaling and availability of ligands. The functions of activins through activin receptors are pleiotrophic, cell type-specific and contextual, and they are involved in the etiology and pathogenesis of a variety of diseases. Accordingly, activin signaling may be a target for therapeutic interventions. In this review, we summarize the current knowledge on activin signaling and discuss the potential roles of this pathway as a molecular target of therapy for metabolic diseases, musculoskeletal disorders, cancers and neural damages.
Collapse
Affiliation(s)
- Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Abnormalities in aggression and anxiety in transgenic mice overexpressing activin E. Biochem Biophys Res Commun 2009; 385:319-23. [PMID: 19463785 DOI: 10.1016/j.bbrc.2009.05.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 05/13/2009] [Indexed: 01/17/2023]
Abstract
To study the function of activin E, a TGF-beta superfamily member, in the regulation of affective behavior, we investigated the behavior of transgenic mice overexpressing activin E (TgActbetaE mice). Male TgActbetaE mice showed aggressive behavior in resident-intruder tests. In elevated plus-maze tests, the percentage of open arm entries was significantly increased in female TgActbetaE mice compared with that in wild-type mice. Furthermore, female TgActbetaE mice stayed in the central area for a significantly longer time than wild-type mice in open field tests. These results indicated that TgActbetaE mice had less anxiety-like behavior. The number of restraint-stress-evoked c-Fos-positive cells in the hypothalamic paraventricular nucleus in TgActbetaE mice was significantly decreased compared with that in wild-type mice. This suggests that synthesis of corticotrophin-releasing hormone induced by stress was decreased in TgActbetaE mice. Taking these results together, activin E may act as a regulator of the hypothalamic-pituitary-adrenal axis.
Collapse
|
50
|
Musazzi L, Cattaneo A, Tardito D, Barbon A, Gennarelli M, Barlati S, Racagni G, Popoli M. Early raise of BDNF in hippocampus suggests induction of posttranscriptional mechanisms by antidepressants. BMC Neurosci 2009; 10:48. [PMID: 19439074 PMCID: PMC2689227 DOI: 10.1186/1471-2202-10-48] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 05/13/2009] [Indexed: 01/01/2023] Open
Abstract
Background The neurotrophin BDNF has been implicated in the regulation of neuroplasticity, gene expression, and synaptic function in the adult brain, as well as in the pathophysiology of neuropsychiatric disorders and the mechanism of action of antidepressants. Antidepressant treatments have been shown to increase the expression of BDNF mRNA, although the changes measured were found to be different depending on various factors. A few studies only have measured levels of BDNF protein after antidepressant treatments, and poor correlation was found between mRNA and protein changes. We studied the time course of expression of BDNF mRNA and protein during drug treatments, in order to elucidate the temporal profile of regulation of this effector and whether mRNA and protein levels correlate. Rat groups were treated for 1, 2 or 3 weeks with fluoxetine or reboxetine; in additional groups drug treatment was followed by a washout week (3+1). Total BDNF mRNA was measured by Real Time PCR, pro- and mature BDNF proteins were measured by Western blot. Results We found that mature BDNF protein is induced more rapidly than mRNA, by both drugs in hippocampus (weeks 1–2) and by reboxetine in prefrontal/frontal cortex (week 1). The temporal profile of BDNF protein expression was largely inconsistent with that of mRNA, which followed the protein induction and reached a peak at week 3. Conclusion These results suggest that BDNF protein is rapidly elevated by antidepressant treatments by posttranscriptional mechanisms, and that induction of BDNF mRNA is a slower process.
Collapse
Affiliation(s)
- Laura Musazzi
- Center of Neuropharmacology-Department of Pharmacological Sciences, University of Milan, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|