1
|
Xin Y, Zhou S, Chu T, Zhou Y, Xu A. Protective Role of Electroacupuncture Against Cognitive Impairment in Neurological Diseases. Curr Neuropharmacol 2025; 23:145-171. [PMID: 38379403 PMCID: PMC11793074 DOI: 10.2174/1570159x22999240209102116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 02/22/2024] Open
Abstract
Many neurological diseases can lead to cognitive impairment in patients, which includes dementia and mild cognitive impairment and thus create a heavy burden both to their families and public health. Due to the limited effectiveness of medications in treating cognitive impairment, it is imperative to develop alternative treatments. Electroacupuncture (EA), a required method for Traditional Chinese Medicine, has the potential treatment of cognitive impairment. However, the molecular mechanisms involved have not been fully elucidated. Considering the current research status, preclinical literature published within the ten years until October 2022 was systematically searched through PubMed, Web of Science, MEDLINE, Ovid, and Embase. By reading the titles and abstracts, a total of 56 studies were initially included. It is concluded that EA can effectively ameliorate cognitive impairment in preclinical research of neurological diseases and induce potentially beneficial changes in molecular pathways, including Alzheimer's disease, vascular cognitive impairment, chronic pain, and Parkinson's disease. Moreover, EA exerts beneficial effects through the same or diverse mechanisms for different disease types, including but not limited to neuroinflammation, neuronal apoptosis, neurogenesis, synaptic plasticity, and autophagy. However, these findings raise further questions that need to be elucidated. Overall, EA therapy for cognitive impairment is an area with great promise, even though more research regarding its detailed mechanisms is warranted.
Collapse
Affiliation(s)
- Yueyang Xin
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Chu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqun Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aijun Xu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Jia F, Han W, Gao S, Huang J, Zhao W, Lu Z, Zhao W, Li Z, Wang Z, Guo Y. Novel cuproptosis metabolism-related molecular clusters and diagnostic signature for Alzheimer's disease. Front Mol Biosci 2024; 11:1478611. [PMID: 39513039 PMCID: PMC11540791 DOI: 10.3389/fmolb.2024.1478611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Background Alzheimer's disease (AD) is a progressive neurodegenerative disorder with no effective treatments available. There is growing evidence that cuproptosis contributes to the pathogenesis of this disease. This study developed a novel molecular clustering based on cuproptosis-related genes and constructed a signature for AD patients. Methods The differentially expressed cuproptosis-related genes (DECRGs) were identified using the DESeq2 R package. The GSEA, PPI network, GO, KEGG, and correlation analysis were conducted to explore the biological functions of DECRGs. Molecular clusters were performed using unsupervised cluster analysis. Differences in biological processes between clusters were evaluated by GSVA and immune infiltration analysis. The optimal model was constructed by WGCNA and machine learning techniques. Decision curve analysis, calibration curves, receiver operating characteristic (ROC) curves, and two additional datasets were employed to confirm the prediction results. Finally, immunofluorescence (IF) staining in AD mice models was used to verify the expression levels of risk genes. Results GSEA and CIBERSORT showed higher levels of resting NK cells, M2 macrophages, naïve CD4+ T cells, neutrophils, monocytes, and plasma cells in AD samples compared to controls. We classified 310 AD patients into two molecular clusters with distinct expression profiles and different immunological characteristics. The C1 subtype showed higher abundance of cuproptosis-related genes, with higher proportions of regulatory T cells, CD8+T cells, and resting dendritic cells. We subsequently constructed a diagnostic model which was confirmed by nomogram, calibration, and decision curve analysis. The values of area under the curves (AUC) were 0.738 and 0.931 for the external datasets, respectively. The expression levels of risk genes were further validated in mouse brain samples. Conclusion Our study provided potential targets for AD treatment, developed a promising gene signature, and offered novel insights for exploring the pathogenesis of AD.
Collapse
Affiliation(s)
- Fang Jia
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wanhong Han
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shuangqi Gao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianwei Huang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wujie Zhao
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhenwei Lu
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenpeng Zhao
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhangyu Li
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhanxiang Wang
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
3
|
Savarimuthu A, Ponniah RJ. Receive, Retain and Retrieve: Psychological and Neurobiological Perspectives on Memory Retrieval. Integr Psychol Behav Sci 2024; 58:303-318. [PMID: 36738400 DOI: 10.1007/s12124-023-09752-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
Memory and learning are interdependent processes that involve encoding, storage, and retrieval. Especially memory retrieval is a fundamental cognitive ability to recall memory traces and update stored memory with new information. For effective memory retrieval and learning, the memory must be stabilized from short-term memory to long-term memory. Hence, it is necessary to understand the process of memory retention and retrieval that enhances the process of learning. Though previous cognitive neuroscience research has focused on memory acquisition and storage, the neurobiological mechanisms underlying memory retrieval and its role in learning are less understood. Therefore, this article offers the viewpoint that memory retrieval is essential for selecting, reactivating, stabilizing, and storing information in long-term memory. In arguing how memories are retrieved, consolidated, transmitted, and strengthened for the long term, the article will examine the psychological and neurobiological aspects of memory and learning with synaptic plasticity, long-term potentiation, genetic transcription, and theta oscillation in the brain.
Collapse
Affiliation(s)
- Anisha Savarimuthu
- Department of Humanities and Social Sciences, National Institute of Technology, Tiruchirappalli, India
| | - R Joseph Ponniah
- Department of Humanities and Social Sciences, National Institute of Technology, Tiruchirappalli, India.
| |
Collapse
|
4
|
Mellios N, Papageorgiou G, Gorgievski V, Maxson G, Hernandez M, Otero M, Varangis M, Dell'Orco M, Perrone-Bizzozero N, Tzavara E. Regulation of neuronal circHomer1 biogenesis by PKA/CREB/ERK-mediated pathways and effects of glutamate and dopamine receptor blockade. RESEARCH SQUARE 2024:rs.3.rs-3547375. [PMID: 38260249 PMCID: PMC10802743 DOI: 10.21203/rs.3.rs-3547375/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
There are currently only very few efficacious drug treatments for SCZ and BD, none of which can significantly ameliorate cognitive symptoms. Thus, further research is needed in elucidating molecular pathways linked to cognitive function and antipsychotic treatment. Circular RNAs (circRNAs) are stable brain-enriched non-coding RNAs, derived from the covalent back-splicing of precursor mRNA molecules. CircHomer1 is a neuronal-enriched, activity-dependent circRNA, derived from the precursor of the long HOMER1B mRNA isoform, which is significantly downregulated in the prefrontal cortex of subjects with psychosis and is able to regulate cognitive function. Even though its relevance to psychiatric disorders and its role in brain function and synaptic plasticity have been well established, little is known about the molecular mechanisms that underlie circHomer1 biogenesis in response to neuronal activity and psychiatric drug treatment. Here we suggest that the RNA-binding protein (RBP) FUS positively regulates neuronal circHomer1 expression. Furthermore, we show that the MEK/ERK and PKA/CREB pathways positively regulate neuronal circHomer1 expression, as well as promote the transcription of Fus and Eif4a3, another RBP previously shown to activate circHomer1 biogenesis. We then demonstrate via both in vitro and in vivo studies that NMDA and mGluR5 receptors are upstream modulators of circHomer1 expression. Lastly, we report that in vivo D2R antagonism increases circHomer1 expression, whereas 5HT2AR blockade reduces circHomer1 levels in multiple brain regions. Taken together, this study allows us to gain novel insights into the molecular circuits that underlie the biogenesis of a psychiatric disease-associated circRNA.
Collapse
|
5
|
Ribeiro FM, Castelo-Branco M, Gonçalves J, Martins J. Visual Cortical Plasticity: Molecular Mechanisms as Revealed by Induction Paradigms in Rodents. Int J Mol Sci 2023; 24:ijms24054701. [PMID: 36902131 PMCID: PMC10003432 DOI: 10.3390/ijms24054701] [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: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
Assessing the molecular mechanism of synaptic plasticity in the cortex is vital for identifying potential targets in conditions marked by defective plasticity. In plasticity research, the visual cortex represents a target model for intense investigation, partly due to the availability of different in vivo plasticity-induction protocols. Here, we review two major protocols: ocular-dominance (OD) and cross-modal (CM) plasticity in rodents, highlighting the molecular signaling pathways involved. Each plasticity paradigm has also revealed the contribution of different populations of inhibitory and excitatory neurons at different time points. Since defective synaptic plasticity is common to various neurodevelopmental disorders, the potentially disrupted molecular and circuit alterations are discussed. Finally, new plasticity paradigms are presented, based on recent evidence. Stimulus-selective response potentiation (SRP) is one of the paradigms addressed. These options may provide answers to unsolved neurodevelopmental questions and offer tools to repair plasticity defects.
Collapse
Affiliation(s)
- Francisco M. Ribeiro
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Joana Gonçalves
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence:
| | - João Martins
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
6
|
Zeng C, Lei D, Lu Y, Huang Q, Wu Y, Yang S, Wu Y. Parvalbumin in the metabolic pathway of glutamate and γ-aminobutyric acid: Influence on expression of GAD65 and GAD67. Arch Biochem Biophys 2023; 734:109499. [PMID: 36587827 DOI: 10.1016/j.abb.2022.109499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022]
Abstract
Parvalbumin-expressing neurons are a type of inhibitory intermediate neuron that play an important role in terminating seizures. The aim of the present study was to use lentiviral construction and packaging technology to overexpress and silence the parvalbumin gene in pheochromocytoma (PC12) cells, and to evaluate how parvalbumin influences the metabolic pathway involving glutamate and γ-aminobutyric acid (GABA). In this work, Immunofluorescence staining was used to verify the differentiation of PC12 cells into neurons after adding nerve growth factor (NGF). Western blotting and real-time quantitative polymerase chain reaction (qRT-PCR) were used to confirm lentivirus-mediated knockdown or overexpression of parvalbumin. Expression of parvalbumin, the 65-kDa GAD isoform (GAD65), and the 67-kDa GAD isoform (GAD67) in neuronal cells was examined at the mRNA and protein levels using qRT-PCR, western blotting and immunofluorescence staining, while intracellular glutamate and GABA levels were determined by high performance liquid chromatography (HPLC). We demonstrate that the expression of parvalbumin is associated with GAD65 and GAD67. Interestingly, overexpression of parvalbumin up-regulated GAD65 and GAD67, increased GABA concentration, and decreased glutamate concentration. Silencing of parvalbumin led to the opposite effects. Altogether, parvalbumin affected the expression of GAD65 and GAD67, thereby influencing the metabolic pathway involving glutamate and GABA.
Collapse
Affiliation(s)
- Chunmei Zeng
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Danqing Lei
- Experimental Center of Life Sciences Institutes, Guangxi Medical University, #22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yuling Lu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Qi Huang
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Ying Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Shengyu Yang
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
7
|
CaMKIV mediates spine growth deficiency of hippocampal neurons by regulation of EGR3/BDNF signal axis in congenital hypothyroidism. Cell Death Dis 2022; 8:482. [PMID: 36473844 PMCID: PMC9723595 DOI: 10.1038/s41420-022-01270-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 11/13/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Congenital hypothyroidism (CH) will cause cognitive impairment in the condition of delayed treatment. The hippocampus is one of the most affected tissues by CH, in which the functional structures of hippocampal neurons manifest deficiency due to aberrant expression of effector molecules. The Ca2+/Calmodulin-dependent protein kinase, CaMKIV, is downregulated in the hippocampal neurons, influencing the growth of dendritic spines in response to CH. However, the underlying mechanism is not fully elucidated. In the present study, the early growth response factor 3 (EGR3) was regulated by CaMKIV in the hippocampal neurons of CH rat pups, as was analyzed by transcriptome sequencing and in vitro cell experiments. EGR3 localized within hippocampal neurons in CA1, CA3, and dentate gyrus regions. Deficient EGR3 in the primary hippocampal neurons significantly reduced the density of dendritic spines by downregulating the expression of BDNF, and such effects could be rescued by supplementing recombinant BDNF protein. Taken together, CH mediates cognitive impairment of pups through the inactivation of CaMKIV in the hippocampal neurons, which decreases the expression of EGR3 and further reduces the production of BDNF, thereby impairing the growth of dendritic spines. Identifying CaMKIV/EGR3/BDNF pathway in the hippocampal neurons in the context of CH will benefit the drug development of intellectual disability caused by CH.
Collapse
|
8
|
Harshini V, Shukla N, Raval I, Kumar S, Shrivastava V, Patel AK, Joshi CG. Kidney transcriptome response to salinity adaptation in Labeo rohita. Front Physiol 2022; 13:991366. [PMID: 36311223 PMCID: PMC9606766 DOI: 10.3389/fphys.2022.991366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
The increasing salinization of freshwater resources, owing to global warming, has caused concern to freshwater aquaculturists. In this regard, the present study is aimed at economically important freshwater fish, L. rohita (rohu) adapting to varying degrees of salinity concentrations. The RNA-seq analysis of kidney tissue samples of L. rohita maintained at 2, 4, 6, and 8 ppt salinity was performed, and differentially expressed genes involved in various pathways were studied. A total of 755, 834, 738, and 716 transcripts were downregulated and 660, 926, 576, and 908 transcripts were up-regulated in 2, 4, 6, and 8 ppt salinity treatment groups, respectively, with reference to the control. Gene ontology enrichment analysis categorized the differentially expressed genes into 69, 154, 92, and 157 numbers of biological processes with the p value < 0.05 for 2, 4, 6, and 8 ppt salinity groups, respectively, based on gene functions. The present study found 26 differentially expressed solute carrier family genes involved in ion transportation and glucose transportation which play a significant role in osmoregulation. In addition, the upregulation of inositol-3-phosphate synthase 1A (INO1) enzyme indicated the role of osmolytes in salinity acclimatization of L. rohita. Apart from this, the study has also found a significant number of genes involved in the pathways related to salinity adaptation including energy metabolism, calcium ion regulation, immune response, structural reorganization, and apoptosis. The kidney transcriptome analysis elucidates a step forward in understanding the osmoregulatory process in L. rohita and their adaptation to salinity changes.
Collapse
Affiliation(s)
- Vemula Harshini
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, India
| | - Nitin Shukla
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, India
| | - Ishan Raval
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, India
| | - Sujit Kumar
- Postgraduate Institute of Fisheries Education and Research, Kamdhenu University, Himmatnagar, Gujarat, India
| | - Vivek Shrivastava
- Postgraduate Institute of Fisheries Education and Research, Kamdhenu University, Himmatnagar, Gujarat, India
| | - Amrutlal K. Patel
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, India
- *Correspondence: Amrutlal K. Patel, ; Chaitanya G. Joshi,
| | - Chaitanya G. Joshi
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, India
- *Correspondence: Amrutlal K. Patel, ; Chaitanya G. Joshi,
| |
Collapse
|
9
|
Jyoti Dutta B, Singh S, Seksaria S, Das Gupta G, Bodakhe SH, Singh A. Potential role of IP3/Ca 2+ signaling and phosphodiesterases: Relevance to neurodegeneration in Alzheimer's disease and possible therapeutic strategies. Biochem Pharmacol 2022; 201:115071. [PMID: 35525328 DOI: 10.1016/j.bcp.2022.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022]
Abstract
Despite large investments by industry and governments, no disease-modifying medications for the treatment of patients with Alzheimer's disease (AD) have been found. The failures of various clinical trials indicate the need for a more in-depth understanding of the pathophysiology of AD and for innovative therapeutic strategies for its treatment. Here, we review the rational for targeting IP3 signaling, cytosolic calcium dysregulation, phosphodiesterases (PDEs), and secondary messengers like cGMP and cAMP, as well as their correlations with the pathophysiology of AD. Various drugs targeting these signaling cascades are still in pre-clinical and clinical trials which support the ideas presented in this article. Further, we describe different molecular mechanisms and medications currently being used in various pre-clinical and clinical trials involving IP3/Ca+2 signaling. We also highlight various isoforms, as well as the functions and pharmacology of the PDEs broadly expressed in different parts of the brain and attempt to unravel the potential benefits of PDE inhibitors for use as novel medications to alleviate the pathogenesis of AD.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur - 495009, Chhattisgarh, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India.
| |
Collapse
|
10
|
κ-Opioid Receptor Agonist Ameliorates Postoperative Neurocognitive Disorder by Activating the Ca 2+/CaMKII/CREB Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3401654. [PMID: 34608407 PMCID: PMC8487382 DOI: 10.1155/2021/3401654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/18/2022]
Abstract
Objective Cardiopulmonary bypass (CPB) is an important cardiac operation and also a high-risk procedure, leading to postoperative neurocognitive disorder. However, there are few effective drugs to treat the aftermath of CPB. Therefore, we observe the effect of kappa opioid receptor (KOR) agonist on cognitive disorders of rats after cardiopulmonary bypass (CPB) and investigate the mechanism of the Ca2+/calmodulin-dependent protein kinase (CaMKII)/cAMP responsive element-binding protein (CREB) pathway. Methods A total of 40 Sprague Dawley rats were randomly divided into the sham operation group (sham group, n = 10), CPB model group (CPB group, n = 10), CPB + KOR agonist U50488H group (UH group, n = 10), and CPB + specific CaMKII antagonist + U50488H group (CKU group, n = 10). The changes in the rats' cognitive function were evaluated using the Morris water maze, the hippocampal histopathological changes were observed via hematoxylin-eosin (H&E) staining, and the apoptosis rate of neuronal cells was detected through terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining. Moreover, enzyme-linked immunosorbent assay (ELISA) was applied to examine the changes in brain injury markers, inflammatory factors, and oxidative stress factors. The hippocampal variations in Ca2+ concentration and oxidative stress index (ROS) levels were measured by immunofluorescence staining, and western blotting was performed to determine the expression changes in the Ca2+/CaMKII/CREB pathway. Results The KOR agonist could shorten latency, increase the swimming distance and residence time in the target quadrant, and ameliorate postoperative neurocognitive disorder (PND). Meanwhile, the KOR agonist relieved CPB-induced hippocampal and oxidative stress injuries, reduced NSE and S-100β expression, decreased the apoptosis rate, and repressed the inflammatory response, which alleviated the brain injury. In addition, U50488H was able to decrease Ca2+ influx and glutamate (Glu) level, inhibit N-methyl-D-aspartate receptor (NMDAR) expression, upregulate CaMKII expression, promote CREB phosphorylation, and increase the brain-derived neurotrophic factor (BDNF) level in CPB rats. However, the protective effects of KORs against PND were suppressed following the application of the CaMKII-specific antagonist. Conclusion The KOR agonist activates the Ca2+/CaMKII/CREB pathway, which improves the brain injury and relieves PND in CPB rats.
Collapse
|
11
|
Wang G, An T, Lei C, Zhu X, Yang L, Zhang L, Zhang R. Antidepressant-like effect of ginsenoside Rb1 on potentiating synaptic plasticity via the miR-134–mediated BDNF signaling pathway in a mouse model of chronic stress-induced depression. J Ginseng Res 2021; 46:376-386. [PMID: 35600767 PMCID: PMC9120625 DOI: 10.1016/j.jgr.2021.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/06/2021] [Accepted: 03/14/2021] [Indexed: 12/28/2022] Open
Abstract
Background Brain-derived neurotrophic factor (BDNF)–tropomyosin-related kinase B (TrkB) plays a critical role in the pathogenesis of depression by modulating synaptic structural remodeling and functional transmission. Previously, we have demonstrated that the ginsenoside Rb1 (Rb1) presents a novel antidepressant-like effect via BDNF–TrkB signaling in the hippocampus of chronic unpredictable mild stress (CUMS)-exposed mice. However, the underlying mechanism through which Rb1 counteracts stress-induced aberrant hippocampal synaptic plasticity via BDNF–TrkB signaling remains elusive. Methods We focused on hippocampal microRNAs (miRNAs) that could directly bind to BDNF and are regulated by Rb1 to explore the possible synaptic plasticity-dependent mechanism of Rb1, which affords protection against CUMS-induced depression-like effects. Results Herein, we observed that brain-specific miRNA-134 (miR-134) could directly bind to BDNF 3′UTR and was markedly downregulated by Rb1 in the hippocampus of CUMS-exposed mice. Furthermore, the hippocampus–targeted miR-134 overexpression substantially blocked the antidepressant-like effects of Rb1 during behavioral tests, attenuating the effects on neuronal nuclei-immunoreactive neurons, the density of dendritic spines, synaptic ultrastructure, long-term potentiation, and expression of synapse-associated proteins and BDNF–TrkB signaling proteins in the hippocampus of CUMS-exposed mice. Conclusion These data provide strong evidence that Rb1 rescued CUMS-induced depression-like effects by modulating hippocampal synaptic plasticity via the miR-134-mediated BDNF signaling pathway. mmu-miR-134-5p could directly bind to BDNF 3’UTR, and was downregulated by Rb1 in the hippocampus of CUMS–exposed mice. miR-134 overexpression blocked the effects of Rb1 on the behavioral tests in CUMS-exposed mice. miR-134 overexpression blocked the effects of Rb1 on synaptic structural changes in the hippocampus of CUMS–exposed mice. miR-134 overexpression blocked the effects of Rb1 on synaptic functional changes in the hippocampus of CUMS–exposed mice. miR-134–mediated BDNF signaling was involved in the antidepressant-like effects of Rb1 in the CUMS–exposed mice.
Collapse
|
12
|
Imoto T, Minoshima M, Yokoyama T, Emery BP, Bull SD, Bito H, Kikuchi K. A Photodeactivatable Antagonist for Controlling CREB-Dependent Gene Expression. ACS CENTRAL SCIENCE 2020; 6:1813-1818. [PMID: 33145417 PMCID: PMC7596873 DOI: 10.1021/acscentsci.0c00736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Indexed: 06/11/2023]
Abstract
A novel photodeactivation strategy for controlling gene expression has been developed based on light-induced activation of cAMP response element binding protein (CREB). Light-induced cleavage of the photoresponsive protecting group of an antagonist of CREB binding protein (CBP) results in photocleaved products with weak binding affinity for CBP. This photodissociation reaction enables protein-protein interactions between CBP and CREB that trigger the formation of a multiprotein transcription complex to turn gene expression "on". This enables irradiation of antagonist-treated HEK293T cells to be used to trigger temporal recovery of CREB-dependent transcriptional activity and endogenous gene expression under photolytic control.
Collapse
Affiliation(s)
- Takuma Imoto
- Division
of Advanced Science and Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masafumi Minoshima
- Division
of Advanced Science and Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tatsushi Yokoyama
- Department
of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ben P. Emery
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Steven D. Bull
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Haruhiko Bito
- Department
of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuya Kikuchi
- Division
of Advanced Science and Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Immunology
Frontier Research Center, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Quantum
Information and Quantum Biology Division, Institute for Open and Transdisciplinary
Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
13
|
Imbe H, Kimura A. Significance of medial preoptic area among the subcortical and cortical areas that are related to pain regulation in the rats with stress-induced hyperalgesia. Brain Res 2020; 1735:146758. [PMID: 32135148 DOI: 10.1016/j.brainres.2020.146758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/24/2020] [Accepted: 02/29/2020] [Indexed: 02/04/2023]
Abstract
Psychophysical stresses frequently increase sensitivity and response to pain, which is termed stress-induced hyperalgesia (SIH). However, the mechanism remains unknown. The subcortical areas such as medial preoptic area (MPO), dorsomedial nucleus of the hypothalamus (DMH), basolateral (BLA) and central nuclei of the amygdala (CeA), and the cortical areas such as insular (IC) and anterior cingulate cortices (ACC) play an important role in pain control via the descending pain modulatory system. In the present study we examined the expression of phosphorylated -cAMP-response element binding protein (pCREB) and the acetylation of histone H3 in these subcortical and cortical areas after repeated restraint stress to reveal changes in the subcortical and cortical areas that affect the function of descending pain modulatory system in the rats with SIH. The repeated restraint stress for 3 weeks induced a decrease in mechanical threshold in the rat hindpaw, an increase in the expression of pCREB in the MPO and an increase in the acetylation of histone H3 in the MPO, BLA and IC. The MPO was the only area that showed an increase in both the expression of pCREB and the acetylation of histone H3 among these examined areas after the repeated restraint stress. Furthermore, the number of pCREB-IR or acetylated histone H3-IR cells in the MPO was negatively correlated with the mechanical threshold. Together, our data represent the importance of the MPO among the subcortical and cortical areas that control descending pain modulatory system under the condition of SIH.
Collapse
Affiliation(s)
- Hiroki Imbe
- Department of Physiology, Wakayama Medical University, Kimiidera 811-1, Wakayama City 641-8509, Japan.
| | - Akihisa Kimura
- Department of Physiology, Wakayama Medical University, Kimiidera 811-1, Wakayama City 641-8509, Japan
| |
Collapse
|
14
|
Fan X, Yan C, Ma Y, Li L, Zhang M, Zhan J. Effect and mechanism of ginsenoside Rg1 on synaptic plasticity of oxygen-glucose deprivation/reoxygenation-induced neuronal injury. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_541_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
15
|
A dual role for peripheral GDNF signaling in nociception and cardiovascular reflexes in the mouse. Proc Natl Acad Sci U S A 2019; 117:698-707. [PMID: 31848242 DOI: 10.1073/pnas.1910905116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Group III/IV muscle afferents transduce nociceptive signals and modulate exercise pressor reflexes (EPRs). However, the mechanisms governing afferent responsiveness to dually modulate these processes are not well characterized. We and others have shown that ischemic injury can induce both nociception-related behaviors and exacerbated EPRs in the same mice. This correlated with primary muscle afferent sensitization and increased expression of glial cell line-derived neurotrophic factor (GDNF) in injured muscle and increased expression of GDNF family receptor α1 (GFRα1) in dorsal root ganglia (DRG). Here, we report that increased GDNF/GFRα1 signaling to sensory neurons from ischemia/reperfusion-affected muscle directly modulated nociceptive-like behaviors and increased exercise-mediated reflexes and group III/IV muscle afferent sensitization. This appeared to have taken effect through increased cyclic adenosine monophosphate (cAMP) response element binding (CREB)/CREB binding protein-mediated expression of the purinergic receptor P2X5 in the DRGs. Muscle GDNF signaling to neurons may, therefore, play an important dual role in nociception and sympathetic reflexes and could provide a therapeutic target for treating complications from ischemic injuries.
Collapse
|
16
|
Liu YY, Brent GA. Thyroid hormone and the brain: Mechanisms of action in development and role in protection and promotion of recovery after brain injury. Pharmacol Ther 2018; 186:176-185. [PMID: 29378220 DOI: 10.1016/j.pharmthera.2018.01.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Thyroid hormone (TH) is essential for normal brain development and may also promote recovery and neuronal regeneration after brain injury. TH acts predominantly through the nuclear receptors, TH receptor alpha (THRA) and beta (THRB). Additional factors that impact TH action in the brain include metabolism, activation of thyroxine (T4) to triiodothyronine (T3) by the enzyme 5'-deiodinase Type 2 (Dio2), inactivation by the enzyme 5-deiodinase Type 3 (Dio3) to reverse T3 (rT3), which occurs in glial cells, and uptake by the Mct8 transporter in neurons. Traumatic brain injury (TBI) is associated with inflammation, metabolic alterations and neural death. In clinical studies, central hypothyroidism, due to hypothalamic and pituitary dysfunction, has been found in some individuals after brain injury. TH has been shown, in animal models, to be protective for the damage incurred from brain injury and may have a role to limit injury and promote recovery. Although clinical trials have not yet been reported, findings from in vitro and in vivo models inform potential treatment strategies utilizing TH for protection and promotion of recovery after brain injury.
Collapse
Affiliation(s)
- Yan-Yun Liu
- Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States
| | - Gregory A Brent
- Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States.
| |
Collapse
|
17
|
Uddin R, Singh SM. Gene Network Construction from Microarray Data Identifies a Key Network Module and Several Candidate Hub Genes in Age-Associated Spatial Learning Impairment. Front Syst Neurosci 2017; 11:75. [PMID: 29066959 PMCID: PMC5641338 DOI: 10.3389/fnsys.2017.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/22/2017] [Indexed: 01/06/2023] Open
Abstract
As humans age many suffer from a decrease in normal brain functions including spatial learning impairments. This study aimed to better understand the molecular mechanisms in age-associated spatial learning impairment (ASLI). We used a mathematical modeling approach implemented in Weighted Gene Co-expression Network Analysis (WGCNA) to create and compare gene network models of young (learning unimpaired) and aged (predominantly learning impaired) brains from a set of exploratory datasets in rats in the context of ASLI. The major goal was to overcome some of the limitations previously observed in the traditional meta- and pathway analysis using these data, and identify novel ASLI related genes and their networks based on co-expression relationship of genes. This analysis identified a set of network modules in the young, each of which is highly enriched with genes functioning in broad but distinct GO functional categories or biological pathways. Interestingly, the analysis pointed to a single module that was highly enriched with genes functioning in “learning and memory” related functions and pathways. Subsequent differential network analysis of this “learning and memory” module in the aged (predominantly learning impaired) rats compared to the young learning unimpaired rats allowed us to identify a set of novel ASLI candidate hub genes. Some of these genes show significant repeatability in networks generated from independent young and aged validation datasets. These hub genes are highly co-expressed with other genes in the network, which not only show differential expression but also differential co-expression and differential connectivity across age and learning impairment. The known function of these hub genes indicate that they play key roles in critical pathways, including kinase and phosphatase signaling, in functions related to various ion channels, and in maintaining neuronal integrity relating to synaptic plasticity and memory formation. Taken together, they provide a new insight and generate new hypotheses into the molecular mechanisms responsible for age associated learning impairment, including spatial learning.
Collapse
Affiliation(s)
- Raihan Uddin
- Department of Biology, University of Western Ontario, London, ON, Canada
| | - Shiva M Singh
- Department of Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
18
|
Electromagnetic Fields for the Regulation of Neural Stem Cells. Stem Cells Int 2017; 2017:9898439. [PMID: 28932245 PMCID: PMC5592400 DOI: 10.1155/2017/9898439] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 08/02/2017] [Indexed: 01/25/2023] Open
Abstract
Localized magnetic fields (MFs) could easily penetrate the scalp, skull, and meninges, thus inducing an electrical current in both the central and peripheral nervous systems, which is primarily used in transcranial magnetic stimulation (TMS) for inducing specific effects on different regions or cells that play roles in various brain activities. Studies of repetitive transcranial magnetic stimulation (rTMS) have led to novel attractive therapeutic approaches. Neural stem cells (NSCs) in adult human brain are able to self-renew and possess multidifferential ability to maintain homeostasis and repair damage after acute central nervous system. In the present review, we summarized the electrical activity of NSCs and the fundamental mechanism of electromagnetic fields and their effects on regulating NSC proliferation, differentiation, migration, and maturation. Although it was authorized for the rTMS use in resistant depression patients by US FDA, there are still unveiling mechanism and limitations for rTMS in clinical applications of acute central nervous system injury, especially on NSC regulation as a rehabilitation strategy. More in-depth studies should be performed to provide detailed parameters and mechanisms of rTMS in further studies, making it a powerful tool to treat people who are surviving with acute central nervous system injuries.
Collapse
|
19
|
Gandolfi D, Cerri S, Mapelli J, Polimeni M, Tritto S, Fuzzati-Armentero MT, Bigiani A, Blandini F, Mapelli L, D'Angelo E. Activation of the CREB/ c-Fos Pathway during Long-Term Synaptic Plasticity in the Cerebellum Granular Layer. Front Cell Neurosci 2017; 11:184. [PMID: 28701927 PMCID: PMC5487453 DOI: 10.3389/fncel.2017.00184] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/16/2017] [Indexed: 12/22/2022] Open
Abstract
The induction of long-term potentiation and depression (LTP and LTD) is thought to trigger gene expression and protein synthesis, leading to consolidation of synaptic and neuronal changes. However, while LTP and LTD have been proposed to play important roles for sensori-motor learning in the cerebellum granular layer, their association with these mechanisms remained unclear. Here, we have investigated phosphorylation of the cAMP-responsive element binding protein (CREB) and activation of the immediate early gene c-Fos pathway following the induction of synaptic plasticity by theta-burst stimulation (TBS) in acute cerebellar slices. LTP and LTD were localized using voltage-sensitive dye imaging (VSDi). At two time points following TBS (15 min and 120 min), corresponding to the early and late phases of plasticity, slices were fixed and processed to evaluate CREB phosphorylation (P-CREB) and c-FOS protein levels, as well as Creb and c-Fos mRNA expression. High levels of P-CREB and Creb/c-Fos were detected before those of c-FOS, as expected if CREB phosphorylation triggered gene expression followed by protein synthesis. No differences between control slices and slices stimulated with TBS were observed in the presence of an N-methyl-D-aspartate receptor (NMDAR) antagonist. Interestingly, activation of the CREB/c-Fos system showed a relevant degree of colocalization with long-term synaptic plasticity. These results show that NMDAR-dependent plasticity at the cerebellum input stage bears about transcriptional and post-transcriptional processes potentially contributing to cerebellar learning and memory consolidation.
Collapse
Affiliation(s)
- Daniela Gandolfi
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of PaviaPavia, Italy.,Brain Connectivity Center, Fondazione Istituto Neurologico Nazionale Casimiro Mondino (IRCCS)Pavia, Italy
| | - Silvia Cerri
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, Fondazione Istituto Neurologico Nazionale Casimiro Mondino (IRCCS)Pavia, Italy
| | - Jonathan Mapelli
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of PaviaPavia, Italy.,Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio EmiliaModena, Italy
| | - Mariarosa Polimeni
- Department of Public Health, Experimental and Forensic Medicine, Human Anatomy Unit, University of PaviaPavia Italy
| | - Simona Tritto
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of PaviaPavia, Italy
| | - Marie-Therese Fuzzati-Armentero
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, Fondazione Istituto Neurologico Nazionale Casimiro Mondino (IRCCS)Pavia, Italy
| | - Albertino Bigiani
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio EmiliaModena, Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, Fondazione Istituto Neurologico Nazionale Casimiro Mondino (IRCCS)Pavia, Italy
| | - Lisa Mapelli
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of PaviaPavia, Italy.,Museo Storico Della Fisica e Centro Studi e Ricerche Enrico FermiRome, Italy
| | - Egidio D'Angelo
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of PaviaPavia, Italy.,Brain Connectivity Center, Fondazione Istituto Neurologico Nazionale Casimiro Mondino (IRCCS)Pavia, Italy
| |
Collapse
|
20
|
Navarro G, Martínez-Pinilla E, Sánchez-Melgar A, Ortiz R, Noé V, Martín M, Ciudad C, Franco R. A genomics approach identifies selective effects of trans-resveratrol in cerebral cortex neuron and glia gene expression. PLoS One 2017; 12:e0176067. [PMID: 28441400 PMCID: PMC5404873 DOI: 10.1371/journal.pone.0176067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/05/2017] [Indexed: 01/28/2023] Open
Abstract
The mode of action of trans-resveratrol, a promising lead compound for the development of neuroprotective drugs, is unknown. Data from a functional genomics study were retrieved with the aim to find differentially expressed genes that may be involved in the benefits provided by trans-resveratrol. Genes that showed a significantly different expression (p<0.05, cut-off of a two-fold change) in mice fed with a control diet or a control diet containing trans-resveratrol were different in cortex, heart and skeletal muscle. In neocortex, we identified 4 up-regulated (Strap, Pkp4, Rab2a, Cpne3) and 22 down-regulated (Actn1, Arf3, Atp6v01, Atp1a3, Atp1b2, Cacng7, Crtc1, Dbn1, Dnm1, Epn1, Gfap, Hap, Mark41, Rab5b, Nrxn2, Ogt, Palm, Ptprn2, Ptprs, Syn2, Timp2, Vamp2) genes upon trans-resveratrol consumption. Network analysis of gene products provided evidence of plakophilin 4 up-regulation as a triggering factor for down-regulation of events related to synaptic vesicle transport and neurotransmitter release via underexpression of dynamin1 and Vamp2 (synaptobrevin 2) as node-gene drivers. Analysis by RT-qPCR of some of the selected genes in a glioma cell line showed that dynamin 1 mRNA was down-regulated even in acute trans-resveratrol treatments. Taken all together, these results give insight on the glial-neuronal networks involved in the neuroprotective role of trans-resveratrol.
Collapse
Affiliation(s)
- Gemma Navarro
- CIBERNED. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Eva Martínez-Pinilla
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, Asturias, Spain
- * E-mail:
| | - Alejandro Sánchez-Melgar
- Facultad de Ciencias y Tecnologías Químicas & Facultad de Medicina. Departamento de Química Inorgánica, Orgánica y Bioquímica, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Raquel Ortiz
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Véronique Noé
- Department of Biochemistry and Physiology, School of Pharmacy, University of Barcelona, Barcelona, Spain
- Institute of Nanotechnology of the University of Barcelona (IN2UB), Barcelona, Spain
| | - Mairena Martín
- Facultad de Ciencias y Tecnologías Químicas & Facultad de Medicina. Departamento de Química Inorgánica, Orgánica y Bioquímica, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Carlos Ciudad
- Department of Biochemistry and Physiology, School of Pharmacy, University of Barcelona, Barcelona, Spain
- Institute of Nanotechnology of the University of Barcelona (IN2UB), Barcelona, Spain
| | - Rafael Franco
- CIBERNED. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Boyce AKJ, Epp AL, Nagarajan A, Swayne LA. Transcriptional and post-translational regulation of pannexins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:72-82. [PMID: 28279657 DOI: 10.1016/j.bbamem.2017.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Pannexins are a 3-membered family of proteins that form large pore ion and metabolite channels in vertebrates. The impact of pannexins on vertebrate biology is intricately tied to where and when they are expressed, and how they are modified, once produced. The purpose of this review is therefore to outline our current understanding of transcriptional and post-translational regulation of pannexins. First, we briefly summarize their discovery and characteristics. Next, we describe several aspects of transcriptional regulation, including cell and tissue-specific expression, dynamic expression over development and disease, as well as new insights into the underlying molecular machinery involved. Following this, we delve into the role of post-translational modifications in the regulation of trafficking and channel properties, highlighting important work on glycosylation, phosphorylation, S-nitrosylation and proteolytic cleavage. Embedded throughout, we also highlight important knowledge gaps and avenues of future research. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Andrew K J Boyce
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Anna L Epp
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Archana Nagarajan
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences and Island Medical Program, University of Victoria, Victoria V8P 5C2, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver V6T 1Z3, Canada.
| |
Collapse
|
22
|
Belgacem YH, Borodinsky LN. CREB at the Crossroads of Activity-Dependent Regulation of Nervous System Development and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:19-39. [PMID: 29080019 DOI: 10.1007/978-3-319-62817-2_2] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The central nervous system is a highly plastic network of cells that constantly adjusts its functions to environmental stimuli throughout life. Transcription-dependent mechanisms modify neuronal properties to respond to external stimuli regulating numerous developmental functions, such as cell survival and differentiation, and physiological functions such as learning, memory, and circadian rhythmicity. The discovery and cloning of the cyclic adenosine monophosphate (cAMP) responsive element binding protein (CREB) constituted a big step toward deciphering the molecular mechanisms underlying neuronal plasticity. CREB was first discovered in learning and memory studies as a crucial mediator of activity-dependent changes in target gene expression that in turn impose long-lasting modifications of the structure and function of neurons. In this chapter, we review the molecular and signaling mechanisms of neural activity-dependent recruitment of CREB and its cofactors. We discuss the crosstalk between signaling pathways that imprints diverse spatiotemporal patterns of CREB activation allowing for the integration of a wide variety of stimuli.
Collapse
Affiliation(s)
- Yesser H Belgacem
- INMED, Aix-Marseille Univ, INSERM, Marseille, France and Aix-Marseille Université, IMéRA, F-13000, Marseille, France.
| | - Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, University of California Davis School of Medicine and Shriners Hospital for Children Northern California, Sacramento, CA, USA
| |
Collapse
|
23
|
Kim J, Lee S, Kang S, Kim SH, Kim JC, Yang M, Moon C. Brain-derived neurotropic factor and GABAergic transmission in neurodegeneration and neuroregeneration. Neural Regen Res 2017; 12:1733-1741. [PMID: 29171440 PMCID: PMC5696856 DOI: 10.4103/1673-5374.217353] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurotoxicity induced by stress, radiation, chemicals, or metabolic diseases, is commonly associated with excitotoxicity, oxidative stress, and neuroinflammation. The pathological process of neurotoxicity induces neuronal death, interrupts synaptic plasticity in the brain, and is similar to that of diverse neurodegenerative diseases. Animal models of neurotoxicity have revealed that clinical symptoms and brain lesions can recover over time via neuroregenerative processes. Specifically, brain-derived neurotropic factor (BDNF) and gamma-aminobutyric acid (GABA)-ergic transmission are related to both neurodegeneration and neuroregeneration. This review summarizes the accumulating evidences that suggest a pathogenic role of BDNF and GABAergic transmission, their underlying mechanisms, and the relationship between BDNF and GABA in neurodegeneration and neuroregeneration. This review will provide a comprehensive overview of the underlying mechanisms of neuroregeneration that may help in developing potential strategies for pharmacotherapeutic approaches to treat neurotoxicity and neurodegenerative disease.
Collapse
Affiliation(s)
- Jinwook Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Sueun Lee
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Sohi Kang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Sung-Ho Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Jong-Choon Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Jeonbuk, South Korea
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
24
|
Abstract
The limited regenerative capacity of neuronal cells requires tight orchestration of cell death and survival regulation in the context of longevity, age-associated diseases as well as during the development of the nervous system. Subordinate to genetic networks epigenetic mechanisms like DNA methylation and histone modifications are involved in the regulation of neuronal development, function and aging. DNA methylation by DNA methyltransferases (DNMTs), mostly correlated with gene silencing, is a dynamic and reversible process. In addition to their canonical actions performing cytosine methylation, DNMTs influence gene expression by interactions with histone modifying enzymes or complexes increasing the complexity of epigenetic transcriptional networks. DNMTs are expressed in neuronal progenitors, post-mitotic as well as adult neurons. In this review, we discuss the role and mode of actions of DNMTs including downstream networks in the regulation of neuronal survival in the developing and aging nervous system and its relevance for associated disorders.
Collapse
Affiliation(s)
- Judit Symmank
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Geraldine Zimmer
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| |
Collapse
|
25
|
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder that has no known cure, nor is there a clear mechanistic understanding of the disease process itself. Although amyloid plaques, neurofibrillary tangles, and cognitive decline are late-stage markers of the disease, it is unclear how they are initially generated, and if they represent a cause, effect, or end phase in the pathology process. Recent studies in AD models have identified marked dysregulations in calcium signaling and related downstream pathways, which occur long before the diagnostic histopathological or cognitive changes. Under normal conditions, intracellular calcium signals are coupled to effectors that maintain a healthy physiological state. Consequently, sustained up-regulation of calcium may have pathophysiological consequences. Indeed, upon reviewing the current body of literature, increased calcium levels are functionally linked to the major features and risk factors of AD: ApoE4 expression, presenilin and APP mutations, beta amyloid plaques, hyperphosphorylation of tau, apoptosis, and synaptic dysfunction. In turn, the histopathological features of AD, once formed, are capable of further increasing calcium levels, leading to a rapid feed-forward acceleration once the disease process has taken hold. The views proposed here consider that AD pathogenesis reflects long-term calcium dysregulations that ultimately serve an enabling role in the disease process. Therefore, “Calcinists” do not necessarily reject βAptist or Tauist doctrine, but rather believe that their genesis is associated with earlier calcium signaling dysregulations. NEUROSCIENTIST 13(5):546—559, 2007.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, IL 60064, USA.
| |
Collapse
|
26
|
Iida T, Yi H, Liu S, Huang W, Kanda H, Lubarsky DA, Hao S. Spinal CPEB-mtROS-CBP signaling pathway contributes to perineural HIV gp120 with ddC-related neuropathic pain in rats. Exp Neurol 2016; 281:17-27. [PMID: 27090160 DOI: 10.1016/j.expneurol.2016.04.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 04/03/2016] [Accepted: 04/12/2016] [Indexed: 10/21/2022]
Abstract
Human immunodeficiency virus (HIV) patients treated with nucleoside reverse transcriptase inhibitors (NRTIs), have been known to develop neuropathic pain. While there has been a major shift away from some neurotoxic NRTIs in current antiretroviral therapy, a large number of HIV patients alive today have previously received them, and many have developed painful peripheral neuropathy. The exact mechanisms by which HIV with NRTIs contribute to the development of neuropathic pain are not known. Previous studies suggest that cytoplasmic polyadenylation element-binding protein (CPEB), reactive oxygen species (ROS), and cAMP-response element-binding protein (CREB)-binding protein (CBP), are involved in the neuroimmunological diseases including inflammatory/neuropathic pain. In this study, we investigated the role of CPEB, mitochondrial ROS (mtROS), or CBP in neuropathic pain induced by HIV envelope protein gp120 combined with antiretroviral drug. The application of recombinant gp120 into the sciatic nerve plus systemic ddC (one of NRTIs) induced mechanical allodynia. Knockdown of CPEB or CBP using intrathecal antisense oligodeoxynucleotide (AS-ODN) reduced mechanical allodynia. Intrathecal mitochondrial superoxide scavenger mito-tempol (Mito-T) increased mechanical withdrawal threshold. Knockdown of CPEB using intrathecal AS-ODN, reduced the up-regulated mitochondrial superoxide in the spinal dorsal horn in rats with gp120 combined with ddC. Intrathecal Mito-T lowered the increased expression of CBP in the spinal dorsal horn. Immunostaining studies showed that neuronal CPEB positive cells were co-localized with MitoSox positive profiles, and that MitoSox positive profiles were co-localized with neuronal CBP. Our studies suggest that neuronal CPEB-mtROS-CBP pathway in the spinal dorsal horn, plays an important role in the gp120/ddC-induced neuropathic pain in rats.
Collapse
Affiliation(s)
- Takafumi Iida
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Department of Anesthesiology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Hyun Yi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Wan Huang
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Hirotsugu Kanda
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Department of Anesthesiology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - David A Lubarsky
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States.
| |
Collapse
|
27
|
Hueston CE, Olsen D, Li Q, Okuwa S, Peng B, Wu J, Volkan PC. Chromatin Modulatory Proteins and Olfactory Receptor Signaling in the Refinement and Maintenance of Fruitless Expression in Olfactory Receptor Neurons. PLoS Biol 2016; 14:e1002443. [PMID: 27093619 PMCID: PMC4836687 DOI: 10.1371/journal.pbio.1002443] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/17/2016] [Indexed: 11/18/2022] Open
Abstract
During development, sensory neurons must choose identities that allow them to detect specific signals and connect with appropriate target neurons. Ultimately, these sensory neurons will successfully integrate into appropriate neural circuits to generate defined motor outputs, or behavior. This integration requires a developmental coordination between the identity of the neuron and the identity of the circuit. The mechanisms that underlie this coordination are currently unknown. Here, we describe two modes of regulation that coordinate the sensory identities of Drosophila melanogaster olfactory receptor neurons (ORNs) involved in sex-specific behaviors with the sex-specific behavioral circuit identity marker fruitless (fru). The first mode involves a developmental program that coordinately restricts to appropriate ORNs the expression of fru and two olfactory receptors (Or47b and Ir84a) involved in sex-specific behaviors. This regulation requires the chromatin modulatory protein Alhambra (Alh). The second mode relies on the signaling from the olfactory receptors through CamK and histone acetyl transferase p300/CBP to maintain ORN-specific fru expression. Our results highlight two feed-forward regulatory mechanisms with both developmentally hardwired and olfactory receptor activity-dependent components that establish and maintain fru expression in ORNs. Such a dual mechanism of fru regulation in ORNs might be a trait of neurons driving plastic aspects of sex-specific behaviors.
Collapse
Affiliation(s)
- Catherine E. Hueston
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
| | - Douglas Olsen
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Qingyun Li
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Sumie Okuwa
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Bo Peng
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Jianni Wu
- Undergraduate Program in Neuroscience, Duke University, Durham, North Carolina, United States of America
| | - Pelin Cayirlioglu Volkan
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- Duke Institute for Brain Science, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
28
|
Nam KN, Mounier A, Fitz NF, Wolfe C, Schug J, Lefterov I, Koldamova R. RXR controlled regulatory networks identified in mouse brain counteract deleterious effects of Aβ oligomers. Sci Rep 2016; 6:24048. [PMID: 27051978 PMCID: PMC4823697 DOI: 10.1038/srep24048] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/16/2016] [Indexed: 11/21/2022] Open
Abstract
Bexarotene, a selective agonist for Retinoid X receptors (RXR) improves cognitive deficits and amyloid-β (Aβ) clearance in mice. Here we examine if the effect of bexarotene on RXR cistrome and transcriptomes depend on APOE isoform and Aβ deposition. We found bexarotene increased RXR binding to promoter regions in cortex of APOE3 mice. Rho family GTPases and Wnt signaling pathway were highly enriched in ChIP-seq and RNA-seq datasets and members of those pathways - Lrp1, Lrp5, Sfrp5 and Sema3f were validated. The effect of APOE isoform was compared in APOE3 and APOE4 mice and we found significant overlapping in affected pathways. ChIP-seq using mouse embryonic stem cells and enrichment levels of histone marks H3K4me3 and H3K27me3 revealed that, bexarotene induced epigenetic changes, consistent with increased neuronal differentiation and in correlation with changes in transcription. Comparison of transcriptome in APOE3 and APP/APOE3 mice revealed that amyloid deposition significantly affects the response to bexarotene. In primary neurons, bexarotene ameliorated the damaged dendrite complexity and loss of neurites caused by Aβ42. Finally, we show that the disruption of actin cytoskeleton induced by Aβ42in vitro was inhibited by bexarotene treatment. Our results suggest a mechanism to establish RXR therapeutic targets with significance in neurodegeneration.
Collapse
Affiliation(s)
- Kyong Nyon Nam
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Anais Mounier
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Nicholas F Fitz
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Cody Wolfe
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Jonathan Schug
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Iliya Lefterov
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Radosveta Koldamova
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| |
Collapse
|
29
|
Horigane SI, Ageta-Ishihara N, Kamijo S, Fujii H, Okamura M, Kinoshita M, Takemoto-Kimura S, Bito H. Facilitation of axon outgrowth via a Wnt5a-CaMKK-CaMKIα pathway during neuronal polarization. Mol Brain 2016; 9:8. [PMID: 26772170 PMCID: PMC4715351 DOI: 10.1186/s13041-016-0189-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/10/2016] [Indexed: 11/10/2022] Open
Abstract
Background Wnt5a, originally identified as a guidance cue for commissural axons, activates a non-canonical pathway critical for cortical axonal morphogenesis. The molecular signaling cascade underlying this event remains obscure. Results Through Ca2+ imaging in acute embryonic cortical slices, we tested if radially migrating cortical excitatory neurons that already bore primitive axons were sensitive to Wnt5a. While Wnt5a only evoked brief Ca2+ transients in immature neurons present in the intermediate zone (IZ), Wnt5a-induced Ca2+ oscillations were sustained in neurons that migrated out to the cortical plate (CP). We wondered whether this early Wnt5a-Ca2+ signaling during neuronal polarization has a morphogenetic consequence. During transition from round to polarized shape, Wnt5a administration to immature cultured cortical neurons specifically promoted axonal, but not dendritic, outgrowth. Pharmacological and genetic inhibition of the CaMKK-CaMKIα pathway abolished Wnt5a-induced axonal elongation, and rescue of CaMKIα in CaMKIα-knockdown neurons restored Wnt5a-mediated axon outgrowth. Conclusions This study suggests that Wnt5a activates Ca2+ signaling during a neuronal morphogenetic time window when axon outgrowth is critically facilitated. Furthermore, the CaMKK-CaMKIα cascade is required for the axonal growth effect of Wnt5a during neuronal polarization. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0189-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shin-ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.
| | - Natsumi Ageta-Ishihara
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Furo-cho, Chikusa, Nagoya, 464-8602, Japan.
| | - Satoshi Kamijo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Michiko Okamura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Furo-cho, Chikusa, Nagoya, 464-8602, Japan.
| | - Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan. .,PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, 102-0076, Japan.
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| |
Collapse
|
30
|
Calcium/calmodulin-dependent protein kinase IV: A multifunctional enzyme and potential therapeutic target. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:54-65. [PMID: 26773169 DOI: 10.1016/j.pbiomolbio.2015.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/21/2015] [Accepted: 12/28/2015] [Indexed: 01/23/2023]
Abstract
The calcium/calmodulin-dependent protein kinase IV (CAMKIV) belongs to the serine/threonine protein kinase family, and is primarily involved in transcriptional regulation in lymphocytes, neurons and male germ cells. CAMKIV operates the signaling cascade and regulates activity of several transcription activators by phosphorylation, which in turn plays pivotal roles in immune response, inflammation and memory consolidation. In this review, we tried to focus on different aspects of CAMKIV to understand the significance of this protein in the biological system. This enzyme is associated with varieties of disorders such as cerebral hypoxia, azoospermia, endometrial and ovarian cancer, systemic lupus, etc., and hence it is considered as a potential therapeutic target. Structure of CAMKIV is comprised of five distinct domains in which kinase domain is responsible for enzyme activity. CAMKIV is involved in varieties of cellular functions such as regulation of gene expression, T-cell maturation, regulation of survival phase of dendritic cells, bone growth and metabolism, memory consolidation, sperm motility, regulation of microtubule dynamics, cell-cycle progression and apoptosis. In this review, we performed an extensive analysis on structure, function and regulation of CAMKIV and associated diseases.
Collapse
|
31
|
Ebbers L, Satheesh SV, Janz K, Rüttiger L, Blosa M, Hofmann F, Morawski M, Griesemer D, Knipper M, Friauf E, Nothwang HG. L-type Calcium Channel Cav1.2 Is Required for Maintenance of Auditory Brainstem Nuclei. J Biol Chem 2015; 290:23692-710. [PMID: 26242732 DOI: 10.1074/jbc.m115.672675] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Indexed: 12/13/2022] Open
Abstract
Cav1.2 and Cav1.3 are the major L-type voltage-gated Ca(2+) channels in the CNS. Yet, their individual in vivo functions are largely unknown. Both channel subunits are expressed in the auditory brainstem, where Cav1.3 is essential for proper maturation. Here, we investigated the role of Cav1.2 by targeted deletion in the mouse embryonic auditory brainstem. Similar to Cav1.3, loss of Cav1.2 resulted in a significant decrease in the volume and cell number of auditory nuclei. Contrary to the deletion of Cav1.3, the action potentials of lateral superior olive (LSO) neurons were narrower compared with controls, whereas the firing behavior and neurotransmission appeared unchanged. Furthermore, auditory brainstem responses were nearly normal in mice lacking Cav1.2. Perineuronal nets were also unaffected. The medial nucleus of the trapezoid body underwent a rapid cell loss between postnatal days P0 and P4, shortly after circuit formation. Phosphorylated cAMP response element-binding protein (CREB), nuclear NFATc4, and the expression levels of p75NTR, Fas, and FasL did not correlate with cell death. These data demonstrate for the first time that both Cav1.2 and Cav1.3 are necessary for neuronal survival but are differentially required for the biophysical properties of neurons. Thus, they perform common as well as distinct functions in the same tissue.
Collapse
Affiliation(s)
- Lena Ebbers
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Somisetty V Satheesh
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Katrin Janz
- the Animal Physiology Group, Department of Biology, University of Kaiserlautern, P. O. Box 3049, 67663 Kaiserslautern, Germany
| | - Lukas Rüttiger
- the Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede Aulhorn Strasse 5, 72076 Tübingen, Germany
| | - Maren Blosa
- the Paul Flechsig Institute of Brain Research, Faculty of Medicine, University Leipzig, Liebigstrasse 19, 04103 Leipzig, Germany
| | - Franz Hofmann
- the Institut für Pharmakologie und Toxikologie, Technische Universität, Biedersteiner Strasse 29, D-80802 München, and
| | - Markus Morawski
- the Paul Flechsig Institute of Brain Research, Faculty of Medicine, University Leipzig, Liebigstrasse 19, 04103 Leipzig, Germany
| | - Désirée Griesemer
- the Animal Physiology Group, Department of Biology, University of Kaiserlautern, P. O. Box 3049, 67663 Kaiserslautern, Germany
| | - Marlies Knipper
- the Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede Aulhorn Strasse 5, 72076 Tübingen, Germany
| | - Eckhard Friauf
- the Animal Physiology Group, Department of Biology, University of Kaiserlautern, P. O. Box 3049, 67663 Kaiserslautern, Germany
| | - Hans Gerd Nothwang
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany, the Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| |
Collapse
|
32
|
Alkadhi KA, Tran TT. Chronic Stress Decreases Basal Levels of Memory-Related Signaling Molecules in Area CA1 of At-Risk (Subclinical) Model of Alzheimer’s Disease. Mol Neurobiol 2015; 52:93-100. [DOI: 10.1007/s12035-014-8839-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/30/2014] [Indexed: 11/28/2022]
|
33
|
Nonaka M, Kim R, Fukushima H, Sasaki K, Suzuki K, Okamura M, Ishii Y, Kawashima T, Kamijo S, Takemoto-Kimura S, Okuno H, Kida S, Bito H. Region-Specific Activation of CRTC1-CREB Signaling Mediates Long-Term Fear Memory. Neuron 2014; 84:92-106. [DOI: 10.1016/j.neuron.2014.08.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2014] [Indexed: 11/29/2022]
|
34
|
Choi YK, Lee DH, Seo YK, Jung H, Park JK, Cho H. Stimulation of Neural Differentiation in Human Bone Marrow Mesenchymal Stem Cells by Extremely Low-Frequency Electromagnetic Fields Incorporated with MNPs. Appl Biochem Biotechnol 2014; 174:1233-1245. [DOI: 10.1007/s12010-014-1091-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/22/2014] [Indexed: 11/30/2022]
|
35
|
Roy A, Modi KK, Khasnavis S, Ghosh S, Watson R, Pahan K. Enhancement of morphological plasticity in hippocampal neurons by a physically modified saline via phosphatidylinositol-3 kinase. PLoS One 2014; 9:e101883. [PMID: 25007337 PMCID: PMC4090203 DOI: 10.1371/journal.pone.0101883] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022] Open
Abstract
Increase of the density of dendritic spines and enhancement of synaptic transmission through ionotropic glutamate receptors are important events, leading to synaptic plasticity and eventually hippocampus-dependent spatial learning and memory formation. Here we have undertaken an innovative approach to upregulate hippocampal plasticity. RNS60 is a 0.9% saline solution containing charge-stabilized nanobubbles that are generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not NS (normal saline), PNS60 (saline containing a comparable level of oxygen without the TCP modification), or RNS10.3 (TCP-modified normal saline without excess oxygen), stimulated morphological plasticity and synaptic transmission via NMDA- and AMPA-sensitive calcium influx in cultured mouse hippocampal neurons. Using mRNA-based targeted gene array, real-time PCR, immunoblot, and immunofluorescence analyses, we further demonstrate that RNS60 stimulated the expression of many plasticity-associated genes in cultured hippocampal neurons. Activation of type IA, but not type IB, phosphatidylinositol-3 (PI-3) kinase by RNS60 together with abrogation of RNS60-mediated upregulation of plasticity-related proteins (NR2A and GluR1) and increase in spine density, neuronal size, and calcium influx by LY294002, a specific inhibitor of PI-3 kinase, suggest that RNS60 upregulates hippocampal plasticity via activation of PI-3 kinase. Finally, in the 5XFAD transgenic model of Alzheimer's disease (AD), RNS60 treatment upregulated expression of plasticity-related proteins PSD95 and NR2A and increased AMPA- and NMDA-dependent hippocampal calcium influx. These results describe a novel property of RNS60 in stimulating hippocampal plasticity, which may help AD and other dementias.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Khushbu K. Modi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Saurabh Khasnavis
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Supurna Ghosh
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Richard Watson
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
36
|
Leone L, Fusco S, Mastrodonato A, Piacentini R, Barbati SA, Zaffina S, Pani G, Podda MV, Grassi C. Epigenetic Modulation of Adult Hippocampal Neurogenesis by Extremely Low-Frequency Electromagnetic Fields. Mol Neurobiol 2014; 49:1472-86. [DOI: 10.1007/s12035-014-8650-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/22/2014] [Indexed: 12/22/2022]
|
37
|
Non-classical effects of estradiol on cAMP responsive element binding protein phosphorylation in gonadotropin-releasing hormone neurons: mechanisms and role. Front Neuroendocrinol 2014; 35:31-41. [PMID: 23978477 DOI: 10.1016/j.yfrne.2013.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/29/2013] [Accepted: 08/12/2013] [Indexed: 12/17/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is produced by a heterogenous neuronal population in the hypothalamus to control pituitary gonadotropin production and reproductive function in all mammalian species. Estradiol is a critical component for the communication between the gonads and the central nervous system. Resolving the mechanisms by which estradiol modulates GnRH neurons is critical for the understanding of how fertility is regulated. Extensive studies during the past decades have provided compelling evidence that estradiol has the potential to alter the intracellular signal transduction mechanisms. The common target of many signaling pathways is the phosphorylation of a key transcription factor, the cAMP response element binding protein (CREB). This review first addresses the aspects of estradiol action on CREB phosphorylation (pCREB) in GnRH neurons. Secondly, this review considers the receptors and signaling network that regulates estradiol's action on pCREB within GnRH neurons and finally it summarizes the physiological significance of CREB to estrogen feedback.
Collapse
|
38
|
Neurogenesis recovery induced by granulocyte-colony stimulating factor in neonatal rat brain after perinatal hypoxia. Pediatr Neonatol 2013; 54:380-8. [PMID: 23791015 DOI: 10.1016/j.pedneo.2013.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 03/14/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Perinatal hypoxia can lead to a wide range of neurological deficits depending on the differential vulnerability of the involved brain regions to oxygen deprivation. It remains unclear whether the differential vulnerability to oxygen deprivation leads to altered neurogenesis in the neonatal brain after perinatal hypoxia. The primary objective was to investigate whether perinatal hypoxia induces deleterious changes in neurogenesis within three representative brain regions (dentate gyrus of the hippocampus, midbrain, and temporal cortex), with regards to common pathological areas clinically. The secondary objective was to investigate whether granulocyte-colony stimulating factor (G-CSF) therapy exerts beneficial effects in neurogenesis in neonatal rat brains subjected to experimental perinatal hypoxia. MATERIALS AND METHODS Rat pups were subjected to experimental perinatal hypoxia on the tenth day of life (P10). They were then given G-CSF (30 μg/kg, single injection/day, intraperitoneal injection, P11-16). The neurogenesis efficacy was analyzed on P17 and the radial-arm maze task, a memory task for higher cognitive functions such as problem-solving abilities, was evaluated on P37-58. RESULTS Perinatal hypoxia caused a significant decrease in neurogenesis within the three representative brain regions, and this deleterious outcome was alleviated by G-CSF (p < 0.05). In addition, the G-CSF therapy markedly improved the decreased performance of long-term cognitive functions induced by perinatal hypoxia (p < 0.05). CONCLUSION This study suggests that G-CSF may be a potentially beneficial therapy, at least in part, through universal recovery of neurogenesis effects in the neonatal brain after perinatal hypoxia insult.
Collapse
|
39
|
Kusuda R, Ravanelli MI, Cadetti F, Franciosi A, Previdelli K, Zanon S, Lucas G. Long-Term Antidepressant Treatment Inhibits Neuropathic Pain-Induced CREB and PLCγ-1 Phosphorylation in the Mouse Spinal Cord Dorsal Horn. THE JOURNAL OF PAIN 2013; 14:1162-72. [DOI: 10.1016/j.jpain.2013.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 04/23/2013] [Accepted: 04/24/2013] [Indexed: 01/10/2023]
|
40
|
Abstract
Adenosine's role in the nervous system is multifaceted. As the core molecule of adenosine triphosphate (ATP), adenosine exists in equilibrium with the adenine nucleotide pool and contributes to cellular energy charge, a quantification of relative amounts of available ATP, ADP, AMP and adenosine. Beyond participating in overall energy balance and thus in maintaining cellular homeostasis, adenosine critically influences dynamic signaling in the nervous system. In particular, adenosine has an effect on and is affected by excitatory synaptic transmission. This report describes the ubiquitous nature of adenosine's influence, outlines specific scenarios of clinical import and highlights emerging knowledge about the regulation of adenosine.
Collapse
Affiliation(s)
- Susan A Masino
- Neuroscience Program and Department of Psychology, 300 Summit St., Trinity College, Hartford, CT 06106, USA.
| | | |
Collapse
|
41
|
Cao J, Wang S, Wang J, Cui W, Nesil T, Vigorito M, Chang SL, Li MD. RNA deep sequencing analysis reveals that nicotine restores impaired gene expression by viral proteins in the brains of HIV-1 transgenic rats. PLoS One 2013; 8:e68517. [PMID: 23874651 PMCID: PMC3712985 DOI: 10.1371/journal.pone.0068517] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 05/06/2013] [Indexed: 01/06/2023] Open
Abstract
Persons infected with HIV-1 often develop neurologic disorders despite receiving highly active anti-retroviral therapy. Although the underlying mechanism is largely undetermined, our previous RNA-seq-based study showed that the expression of many genes was altered in the central nervous system (CNS) of HIV-1 transgenic (HIV-1Tg) rats. Because nicotine, a natural agonist of nicotinic acetylcholine receptors, exhibits a neuroprotective effect, we presently tested the hypothesis that nicotine restores the expression of altered genes in the CNS of HIV-1Tg rats. Adult male HIV-1Tg and F344 control strain rats were injected with either nicotine (0.25 mg/kg) or saline subcutaneously twice a day for 17 days. Gene expression in the prefrontal cortex (PFC), dorsal hippocampus (HIP), and dorsal striatum (STR) was evaluated using the RNA deep sequencing technique. We found that about 20% of the altered genes in the HIV-1Tg rat were affected by nicotine in each brain region, with the expression of most restored. Analysis of the restored genes showed distinct pathways corrected by nicotine in different brain regions of HIV-1Tg rats. Specifically, the two most significantly restored pathways were Wnt/β-catenin signaling and ephrin B signaling in the PFC, cAMP-responsive element-binding protein (CREB) signaling and glutathione metabolism pathway in the HIP, and tricarboxylic acid (TCA) cycle and calcium signaling in the STR. Together, our findings indicate that cholinergic modulators such as nicotine have beneficial effects on HIV-1-induced neurologic deficits.
Collapse
Affiliation(s)
- Junran Cao
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shaolin Wang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ju Wang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Wenyan Cui
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Tanseli Nesil
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael Vigorito
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, United States of America
- Department of Psychology, Seton Hall University, South Orange, New Jersey, United States of America
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, United States of America
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, United States of America
| | - Ming D. Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
42
|
Fujii H, Inoue M, Okuno H, Sano Y, Takemoto-Kimura S, Kitamura K, Kano M, Bito H. Nonlinear decoding and asymmetric representation of neuronal input information by CaMKIIα and calcineurin. Cell Rep 2013; 3:978-87. [PMID: 23602566 DOI: 10.1016/j.celrep.2013.03.033] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 10/26/2022] Open
Abstract
How information encoded in glutamate release rates at individual synapses is converted into biochemical activation patterns of postsynaptic enzymes remains unexplored. To address this, we developed a dual fluorescence resonance energy transfer (FRET) imaging platform and recorded CaMKIIα and calcineurin activities in hippocampal neurons while varying glutamate uncaging frequencies. With little spine morphological change, 5 Hz spine glutamate uncaging strongly stimulated calcineurin, but not CaMKIIα. In contrast, 20 Hz spine glutamate uncaging, which induced spine growth, activated both CaMKIIα and calcineurin with distinct spatiotemporal kinetics. Higher temporal resolution recording in the soma revealed that CaMKIIα activity summed supralinearly and sensed both higher frequency and input number, thus acting as an input frequency/number decoder. In contrast, calcineurin activity summated sublinearly with increasing input number and showed little frequency dependence, thus functioning as an input number counter. These results provide evidence that CaMKIIα and calcineurin are fine-tuned to unique bandwidths and compute input variables in an asymmetric manner.
Collapse
Affiliation(s)
- Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kim JE, Kim DS, Jin Ryu H, Il Kim W, Kim MJ, Won Kim D, Young Choi S, Kang TC. The effect of P2X7 receptor activation on nuclear factor-κB phosphorylation induced by status epilepticus in the rat hippocampus. Hippocampus 2013; 23:500-14. [PMID: 23564500 DOI: 10.1002/hipo.22109] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2013] [Indexed: 11/10/2022]
Abstract
Nuclear factor-kappa B (NFκB) signal is essential for neuronal survival and its activation may protect neuron against various stimuli. Since purinergic signals activate NFκB through the P2X7 receptor, we investigated the distinct pattern of NF-κB phosphorylation in neurons by P2X7 receptor activation following status epilepticus (SE) in an effort to understand the role of P2X7 receptor in epileptogenic insult. In non-SE animals, 2'(3')-O-(4-benzoyl)benzoyl adenosine 5'-triphosphate (BzATP, a P2X7R agonist) treatment increased only p52-Ser869 NF-κB phosphorylation in neuron. Following SE, p52-Ser865, p52-Ser869, p65-Ser276, p65-Ser311, p65-Ser468, and p65-Ser529 NF-κB phosphorylation was significantly decreased in CA1 and CA3 neurons. However, BzATP treatment prevented reductions in p65-Ser276, p65-Ser311, p65-Ser529, and p52-Ser869 NF-κB phosphorylations in CA1 and/or CA3 neurons induced by SE. Furthermore, BzATP treatment reduced SE-induced p65-Ser311, p65-Ser468, p65-Ser536, and p52-Ser869 NF-κB phosphorylations in astrocytes. These findings indicate that P2X7 functions may be involved in the regulation of SE-induced reactive astrocytes and neuronal degeneration via NF-κB phosphorylations in response to pilocarpine-induced SE in the rat hippocampus.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Melanocortin-4 receptor regulates hippocampal synaptic plasticity through a protein kinase A-dependent mechanism. J Neurosci 2013; 33:464-72. [PMID: 23303927 DOI: 10.1523/jneurosci.3282-12.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Learning and memory require orchestrated regulation of both structural and functional synaptic plasticity in the hippocampus. While a neuropeptide alpha-melanocyte-stimulating hormone, α-MSH, has been implicated in memory acquisition and retention, the functional role of its cognate receptor, melanocortin-4 receptor (MC4R), in hippocampal-dependent synaptic plasticity has not been explored. In this study, we report that activation of MC4R enhances synaptic plasticity through the regulation of dendritic spine morphology and abundance of AMPA receptors. We show that activation of postsynaptic MC4R increases the number of mature dendritic spines and enhances surface expression of AMPA receptor subunit GluA1, resulting in synaptic accumulation of GluA1-containing AMPA receptors. Moreover, MC4R stimulates surface GluA1 trafficking through phosphorylation of GluA1 at Ser845 in a Gα(s)-cAMP/PKA-dependent manner. Blockade of protein kinase A (PKA) signaling abolishes the MC4R-mediated enhancement of neurotransmission and hippocampal long-term potentiation. Importantly, in vivo application of MC4R agonists increases LTP in the mouse hippocampal CA1 region. These findings reveal that MC4R in the hippocampus plays a critical role in the regulation of structural and functional plasticity.
Collapse
|
45
|
Zara S, De Colli M, Rapino M, Pacella S, Nasuti C, Sozio P, Di Stefano A, Cataldi A. Ibuprofen and lipoic acid conjugate neuroprotective activity is mediated by Ngb/Akt intracellular signaling pathway in Alzheimer's disease rat model. Gerontology 2013; 59:250-60. [PMID: 23428737 DOI: 10.1159/000346445] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 12/11/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a frequent form of senile dementia. Neuroglobin (Ngb) has a neuroprotective role and decreases Aβ peptide levels. Ngb, promoting Akt phosphorylation, activates cell survival involving cyclic-nucleotide response element-binding protein (CREB). A new molecule (IBU-LA) was synthetized and administered to an AD rat model to counteract AD progression. OBJECTIVE The aim of this study was to investigate the IBU-LA-mediated induction of Ngb neuroprotective and antiapoptotic activities. METHODS Brain morphology was analyzed through Bielschowsky staining, Aβ(1-40) and Ngb expression by immunohistochemistry. Akt, p-Akt, CREB and p-CREB expression was evaluated by Western blot, apoptosis through cytochrome C/Apaf 1 immunocomplex formation, and TUNEL analysis. RESULTS Bielschowsky staining and Aβ(1-40) expression show few nerve connections and Aβ(1-40) expression in an Aβ sample, preserved neuronal cells and Aβ(1-40) expression lowering in an IBU sample, mostly in IBU-LA. The Ngb level decreases in Aβ samples, compared to control and IBU-LA samples. p-Akt/Akt and p-CREB/CREB ratios reveal a reduction in Aβ sample, going back to the basal level in control and IBU-LA samples. Cytochrome C/Apaf 1 co-immunoprecipitate occurs and TUNEL-positive nuclei percentage decreases in Aβ sample. Probe test performance shows an increased spatial reference memory in the IBU-LA compared to the Aβ sample; no significant differences were seen between the IBU-LA and IBU samples. CONCLUSION This evidence reveals that IBU-LA administration has the capability to maintain a high Ngb level allowing Ngb to perform a neuroprotective and antiapoptotic role, representing a valid tool in the therapeutic strategy of AD progression.
Collapse
Affiliation(s)
- Susi Zara
- Dipartimento di Farmacia, Università 'G. d'Annunzio', Chieti, Italia.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Ho L, Ferruzzi MG, Janle EM, Wang J, Gong B, Chen TY, Lobo J, Cooper B, Wu QL, Talcott ST, Percival SS, Simon JE, Pasinetti GM. Identification of brain-targeted bioactive dietary quercetin-3-O-glucuronide as a novel intervention for Alzheimer's disease. FASEB J 2013; 27:769-81. [PMID: 23097297 PMCID: PMC3545533 DOI: 10.1096/fj.12-212118] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 10/09/2012] [Indexed: 12/25/2022]
Abstract
Epidemiological and preclinical studies indicate that polyphenol intake from moderate consumption of red wines may lower the relative risk for developing Alzheimer's disease (AD) dementia. There is limited information regarding the specific biological activities and cellular and molecular mechanisms by which wine polyphenolic components might modulate AD. We assessed accumulations of polyphenols in the rat brain following oral dosage with a Cabernet Sauvignon red wine and tested brain-targeted polyphenols for potential beneficial AD disease-modifying activities. We identified accumulations of select polyphenolic metabolites in the brain. We demonstrated that, in comparison to vehicle-control treatment, one of the brain-targeted polyphenol metabolites, quercetin-3-O-glucuronide, significantly reduced the generation of β-amyloid (Aβ) peptides by primary neuron cultures generated from the Tg2576 AD mouse model. Another brain-targeted metabolite, malvidin-3-O-glucoside, had no detectable effect on Aβ generation. Moreover, in an in vitro analysis using the photo-induced cross-linking of unmodified proteins (PICUP) technique, we found that quercetin-3-O-glucuronide is also capable of interfering with the initial protein-protein interaction of Aβ(1-40) and Aβ(1-42) that is necessary for the formation of neurotoxic oligomeric Aβ species. Lastly, we found that quercetin-3-O-glucuronide treatment, compared to vehicle-control treatment, significantly improved AD-type deficits in hippocampal formation basal synaptic transmission and long-term potentiation, possibly through mechanisms involving the activation of the c-Jun N-terminal kinases and the mitogen-activated protein kinase signaling pathways. Brain-targeted quercetin-3-O-glucuronide may simultaneously modulate multiple independent AD disease-modifying mechanisms and, as such, may contribute to the benefits of dietary supplementation with red wines as an effective intervention for AD.
Collapse
Affiliation(s)
- Lap Ho
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | - Jun Wang
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
| | - Bing Gong
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | - Bruce Cooper
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
| | - Qing Li Wu
- Department of Plant Biology and Plant Pathology, Rutgers University, New Brunswick, New Jersey, USA
| | - Stephen T. Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texa, USA; and
| | - Susan S. Percival
- Department of Food Science and Human Nutrition, University of Florida, Gainsville, Florida, USA
| | - James E. Simon
- Department of Plant Biology and Plant Pathology, Rutgers University, New Brunswick, New Jersey, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
- Geriatric Research Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
47
|
Liu B, Barbosa-Sampaio H, Jones PM, Persaud SJ, Muller DS. The CaMK4/CREB/IRS-2 cascade stimulates proliferation and inhibits apoptosis of β-cells. PLoS One 2012; 7:e45711. [PMID: 23049845 PMCID: PMC3458088 DOI: 10.1371/journal.pone.0045711] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/23/2012] [Indexed: 01/09/2023] Open
Abstract
Progressive reduction in β-cell mass is responsible for the development of type 2 diabetes mellitus, and alteration in insulin receptor substrate 2 (IRS-2) abundance plays a critical role in this process. IRS-2 expression is stimulated by the transcription factor cAMP response element-binding protein (CREB) and we recently demonstrated that Ca2+/calmodulin dependent kinase 4 (CaMK4) is upstream of CREB activation in β-cells. This study investigated whether CaMK4 is also a potential target to increase β-cell mass through CREB-mediated IRS-2 expression, by quantifying mouse MIN6 β-cell proliferation and apoptosis following IRS-2 knockdown, CaMKs inhibition and alterations in CaMK4 and CREB expression. Expression of constitutively active CaMK4 (ΔCaMK4) and CREB (CREBDIEDLM) significantly stimulated β-cell proliferation and survival. In contrast, expression of their corresponding dominant negative forms (ΔK75ECaMK4 and CREBM1) and silencing of IRS-2 increased apoptosis and reduced β-cell division. Moreover, CREBDIEDLM and CREBM1 expression completely abolished the effects of ΔK75ECaMK4 and of ΔCaMK4, respectively. Our results indicate that CaMK4 regulates β-cell proliferation and apoptosis in a CREB-dependent manner and that CaMK4-induced IRS-2 expression is important in these processes.
Collapse
Affiliation(s)
- Bo Liu
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| | - Helena Barbosa-Sampaio
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| | - Peter M. Jones
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| | - Shanta J. Persaud
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
- * E-mail:
| | - Dany S. Muller
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
48
|
Fukumitsu H, Soumiya H, Furukawa S. Knockdown of pre-mRNA cleavage factor Im 25 kDa promotes neurite outgrowth. Biochem Biophys Res Commun 2012; 425:848-53. [PMID: 22898046 DOI: 10.1016/j.bbrc.2012.07.164] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/31/2012] [Indexed: 01/16/2023]
Abstract
Mammalian precursor mRNA (pre-mRNA) cleavage factor I (CFIm) plays important roles in the selection of poly(A) sites in a 3'-untranslated region (3'-UTR), producing mRNAs with variable 3' ends. Because 3'-UTRs often contain cis elements that impact stability or localization of mRNA or translation, alternative polyadenylation diversifies utilization of primary transcripts in mammalian cells. However, the physiological role of CFIm remains unclear. CFIm acts as a heterodimer comprising a 25kDa subunit (CFIm25) and one of the three large subunits-CFIm59, CFIm68, or CFIm72. CFIm25 binds directly to RNA and introduces and anchors the larger subunit. To examine the physiological roles of CFIm, we knocked down the CFIm25 gene in neuronal cells using RNA interference. Knockdown of CFIm25 increased the number of primary dendrites of developing hippocampal neurons and promoted nerve growth factor (NGF)-induced neurite extension from rat pheochromocytoma PC12 cells without affecting the morphology of proliferating PC12 cells. On the other hand, CFIm25 knockdown did not influence constitutively active or dominantly negative RhoA suppression or promotion of NGF-induced neurite extension from PC12 cells, respectively. Taken together, our results indicate that endogenous CFIm may promote neuritogenesis in developing neurons by coordinating events upstream of NGF-induced RhoA inactivation.
Collapse
Affiliation(s)
- Hidefumi Fukumitsu
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, Daigakunishi 1-25-4, Gifu 501 1196, Japan.
| | | | | |
Collapse
|
49
|
Lazarovici P, Cohen G, Arien-Zakay H, Chen J, Zhang C, Chopp M, Jiang H. Multimodal neuroprotection induced by PACAP38 in oxygen-glucose deprivation and middle cerebral artery occlusion stroke models. J Mol Neurosci 2012; 48:526-40. [PMID: 22678884 DOI: 10.1007/s12031-012-9818-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 05/21/2012] [Indexed: 01/30/2023]
Abstract
Pituitary adenylate cyclase activating peptide (PACAP), a potent neuropeptide which crosses the blood-brain barrier, is known to provide neuroprotection in rat stroke models of middle cerebral artery occlusion (MCAO) by mechanism(s) which deserve clarification. We confirmed that following i.v. injection of 30 ng/kg of PACAP38 in rats exposed to 2 h of MCAO focal cerebral ischemia and 48 h reoxygenation, 50 % neuroprotection was measured by reduced caspase-3 activity and volume of cerebral infarction. Similar neuroprotective effects were measured upon PACAP38 treatment of oxygen-glucose deprivation and reoxygenation of brain cortical neurons. The neuroprotection was temporally associated with increased expression of brain-derived neurotrophic factor, phosphorylation of its receptor-tropomyosin-related kinase receptor type B (trkB), activation of phosphoinositide 3-kinase and Akt, and reduction of extracellular signal-regulated kinases 1/2 phosphorylation. PACAP38 increased expression of neuronal markers beta-tubulin III, microtubule-associated protein-2, and growth-associated protein-43. PACAP38 induced stimulation of Rac and suppression of Rho GTPase activities. PACAP38 downregulated the nerve growth factor receptor (p75(NTR)) and associated Nogo-(Neurite outgrowth-A) receptor. Collectively, these in vitro and in vivo results propose that PACAP exhibits neuroprotective effects in cerebral ischemia by three mechanisms: a direct one, mediated by PACAP receptors, and two indirect, induced by neurotrophin release, activation of the trkB receptors and attenuation of neuronal growth inhibitory signaling molecules p75(NTR) and Nogo receptor.
Collapse
Affiliation(s)
- Philip Lazarovici
- School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem, POB 12065, Jerusalem 91120, Israel.
| | | | | | | | | | | | | |
Collapse
|
50
|
Bjorklund NL, Reese LC, Sadagoparamanujam VM, Ghirardi V, Woltjer RL, Taglialatela G. Absence of amyloid β oligomers at the postsynapse and regulated synaptic Zn2+ in cognitively intact aged individuals with Alzheimer's disease neuropathology. Mol Neurodegener 2012; 7:23. [PMID: 22640423 PMCID: PMC3403985 DOI: 10.1186/1750-1326-7-23] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 05/28/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early cognitive impairment in Alzheimer Disease (AD) is thought to result from the dysfunctional effect of amyloid beta (Aβ) oligomers targeting the synapses. Some individuals, however, escape cognitive decline despite the presence of the neuropathologic features of AD (Aβ plaques and neurofibrillary tangles). We term this group Non-Demented with AD Neuropathology or NDAN. The present study illustrates one putative resistance mechanism involved in NDAN cases which may suggest targets for the effective treatment of AD. RESULTS Here we describe the localization of Aβ oligomers at the postsynapse in hippocampi from AD cases. Notably, however, we also found that while present in soluble fractions, Aβ oligomers are absent from hippocampal postsynapses in NDAN cases. In addition, levels of phosphorylated (active) CREB, a transcription factor important for synaptic plasticity, are normal in NDAN individuals, suggesting that their synapses are functionally intact. Analysis of Zn2+ showed that levels were increased in both soluble fractions and synaptic vesicles in AD hippocampi, paralleled by a decrease of expression of the synaptic vesicle Zn2+ transporter, ZnT3. Conversely, in NDAN individuals, levels of Zn2+ in soluble fractions were significantly lower than in AD, whereas in synaptic vesicles the levels of Zn2+ were similar to AD, but accompanied by preserved expression of the ZnT3. CONCLUSIONS Taken together, these data illustrate that despite substantial AD neuropathology, Aβ oligomers, and increased synaptic vesicle Zn2+, susceptible brain tissue in these aged NDAN individuals features, as compared to symptomatic AD subjects, significantly lower total Zn2+ levels and no association of Aβ oligomers with the postsynapse, which collectively may promote the maintenance of intact cognitive function.
Collapse
Affiliation(s)
| | - Lindsay C Reese
- Department of Neuroscience and Cell Biology, Galveston, TX, 77555, USA
| | - V-M Sadagoparamanujam
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Valeria Ghirardi
- Department of Neuroscience and Cell Biology, Galveston, TX, 77555, USA
| | - Randall L Woltjer
- Department of Pathology, Oregon Health & Science University, Portland, OR, 97201, USA
| | | |
Collapse
|