1
|
Feng J, Zhao J, Kuang Y, Zhou Y, Ye Z, He Y, Chen D, Zhang L, Zhang T, Zhu Q, Cheng S, Liu T. Hsa_Circ_0105596/FTO inhibits progression of Parkinson's disease by sponging miR-187-3p and regulating eEF2. Heliyon 2024; 10:e39830. [PMID: 39654770 PMCID: PMC11626004 DOI: 10.1016/j.heliyon.2024.e39830] [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: 07/30/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024] Open
Abstract
Background Parkinson's disease (PD) characterized by inflammation and protein erroneous deposition, whose pathological mechanisms have not been elucidated. NcRNA plays important role in PD, especially when circRNA sponges miRNA, which leads to the breakdown of downstream regulation. The aim of this study is to investigate the dynamic changes between upregulated circRNA and downregulated miRNA during the pathogenesis of PD and their impact on downstream miRNA targets. Method We conducted bioinformatics on sequencing data of substantia nigra (SN) and striatum, and intersected differentially expressed genes (Degs) to determine core role of circFTO-miR-187-3p-EEF2 axes in the progression of PD. Firstly, therapeutic effect of knock-down circFTO in PD and its impact on EEF2 were determined in mouse, using immunohistochemistry, HE, Nissl, TUNEL staining and Western blot (WB). Targeted binding relationship between circFTO, miR-187-3p, and EEF2 was determined through RNA binding protein immunoprecipitation assays (RIP) and dual luciferase reporter assay. The significance of gene in apoptosis was confirmed through flow cytometry, lentiviral transduction, quantitative real-time PCR, and WB. Result CircFTO is upregulated and miR-187-3p is downregulated in SN of PD. EEF2 was the core of neural repair related modules in both SN and striatum using Weighted Correlation Network Analysis (WGCNA) and protein-protein interaction (PPI). The binding regulation relationship between circFTO-miR-187-3p-EEF2 was determined through structural analysis, RIP, and dual luciferase reporter assay. After knocking-down circFTO, animals showed alleviated symptoms, decreased levels of oxidative stress and EEF2. Upregulation of miR-187-3p or si-circFTO in SH-SY5Y cells can reduce cell apoptosis, EEF2, and oxidative stress. Moreover, individual interference with EEF2 can partially counteract the induction of 6-OHDA on apoptosis. Conclusion Excessive circFTO sponging miR-187-3p leads to the inability of miR-187-3p to effectively regulate expression of EEF2, resulting in the progression of PD. Moreover, interference with circFTO can effectively reduce brain inflammation and oxidative stress.
Collapse
Affiliation(s)
- Jiahao Feng
- Traditional Chinese Medicine Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Jin Zhao
- Traditional Chinese Medicine Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Yong Kuang
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Yuheng Zhou
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510632, China
| | - Ziheng Ye
- Traditional Chinese Medicine Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Yutong He
- School of medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Dandan Chen
- ChangSha Medical University, Changsha, 41000, China
| | - Li Zhang
- Traditional Chinese Medicine Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Tingying Zhang
- Traditional Chinese Medicine Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Qingqing Zhu
- Scientific Research Center Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Shumin Cheng
- Scientific Research Center Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Taoli Liu
- Traditional Chinese Medicine Department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518000, China
| |
Collapse
|
2
|
Smith PR, Garcia G, Meyer AR, Ryazanov AG, Ma T, Loerch S, Campbell ZT. eEF2K regulates pain through translational control of BDNF. Mol Cell 2024:S1097-2765(24)00948-1. [PMID: 39694034 DOI: 10.1016/j.molcel.2024.11.023] [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: 05/06/2024] [Revised: 10/01/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
mRNA translation is integral to pain, yet the key regulatory factors and their target mRNAs are unclear. Here, we uncover a mechanism that bridges noxious insults to multiple phases of translational control in murine sensory neurons. We find that a painful cue triggers repression of peptide chain elongation through activation of elongation factor 2 kinase (eEF2K). Attenuated elongation is sensed by a ribosome-coupled mechanism that triggers the integrated stress response (ISR). Both eEF2K and the ISR are required for pain-associated behaviors in vivo. This pathway simultaneously induces biosynthesis of brain-derived neurotrophic factor (BDNF). Selective blockade of Bdnf translation has analgesic effects in vivo. Our data suggest that precise spatiotemporal regulation of Bdnf translation is critical for appropriate behavioral responses to painful stimuli. Overall, our results demonstrate that eEF2K resides at the nexus of an intricate regulatory network that links painful cues to multiple layers of translational control.
Collapse
Affiliation(s)
- Patrick R Smith
- Department of Anesthesiology, University of Wisconsin, Madison, Madison, WI, USA
| | - Guadalupe Garcia
- Department of Anesthesiology, University of Wisconsin, Madison, Madison, WI, USA
| | - Angela R Meyer
- Department of Anesthesiology, University of Wisconsin, Madison, Madison, WI, USA
| | - Alexey G Ryazanov
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Tao Ma
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Translational Neuroscience, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sarah Loerch
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA; Center for Molecular Biology of RNA, University of California, Santa Cruz, CA 95064, USA
| | - Zachary T Campbell
- Department of Anesthesiology, University of Wisconsin, Madison, Madison, WI, USA.
| |
Collapse
|
3
|
Kong L, Yang J, Yang H, Xu B, Yang T, Liu W. Research advances on CaMKs-mediated neurodevelopmental injury. Arch Toxicol 2024; 98:3933-3947. [PMID: 39292234 DOI: 10.1007/s00204-024-03865-5] [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: 07/13/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Calcium/calmodulin-dependent protein kinases (CaMKs) are important proteins in the calcium signaling cascade response pathway, which can broadly regulate biological functions in vivo. Multifunctional CaMKs play key roles in neural development, including neuronal circuit building, synaptic plasticity establishment, and neurotrophic factor secretion. Currently, four familial proteins, calcium/calmodulin-dependent protein kinase I (CaMKI), calcium/calmodulin-dependent protein kinase II (CaMKII), eukaryotic elongation factor 2 kinase (eEF2K, popularly known as CaMKIII) and calcium/calmodulin-dependent protein kinase IV (CaMKIV), are thought to have been the most extensively studied during neurodevelopment. Although their spatial structures are extremely similar, as well as the initial starting point of activation, both require the activation of calcium and calmodulin (CaM) complexes to be involved in the process, and the phosphorylation sites and modes of each member are different. Furthermore, due to the high structural similarity of CaMKs, their members may play synergistic roles in the regulation of neural development, but different CaMKs also have their own means of regulating neural development. In this review, we first describe the visualized protein structural forms of CaMKI, CaMKII, eEF2K and CaMKIV, and then describe the functions of each kinase in neurodevelopment. After that, we focus on four main mechanisms of neurodevelopmental damage caused by CaMKs: CaMKI/ERK/CREB pathway inhibition leading to dendritic spine structural damage; Ca2+/CaM/CaMKII through induction of mitochondrial kinetic disorders leading to neurodevelopmental damage; CaMKIII/eEF2 hyperphosphorylation affects the establishment of synaptic plasticity; and CaMKIV/JNK/NF-κB through induction of an inflammatory response leading to neurodevelopmental damage. In conclusion, we briefly discuss the pathophysiological significance of aberrant CaMK family expression in neurodevelopmental disorders, as well as the protective effects of conventional CaMKII and CaMKIII antagonists against neurodevelopmental injury.
Collapse
Affiliation(s)
- Lingxu Kong
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Jing Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Huajie Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Bin Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Tianyao Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China.
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China.
| | - Wei Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China.
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
4
|
Polishchuk A, Cilleros-Mañé V, Balanyà-Segura M, Just-Borràs L, Forniés-Mariné A, Silvera-Simón C, Tomàs M, Jami El Hirchi M, Hurtado E, Tomàs J, Lanuza MA. BDNF/TrkB signalling, in cooperation with muscarinic signalling, retrogradely regulates PKA pathway to phosphorylate SNAP-25 and Synapsin-1 at the neuromuscular junction. Cell Commun Signal 2024; 22:371. [PMID: 39044222 PMCID: PMC11265447 DOI: 10.1186/s12964-024-01735-2] [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: 05/07/2024] [Accepted: 07/04/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Protein kinase A (PKA) enhances neurotransmission at the neuromuscular junction (NMJ), which is retrogradely regulated by nerve-induced muscle contraction to promote Acetylcholine (ACh) release through the phosphorylation of molecules involved in synaptic vesicle exocytosis (SNAP-25 and Synapsin-1). However, the molecular mechanism of the retrograde regulation of PKA subunits and its targets by BDNF/TrkB pathway and muscarinic signalling has not been demonstrated until now. At the NMJ, retrograde control is mainly associated with BDNF/TrkB signalling as muscle contraction enhances BDNF levels and controls specific kinases involved in the neurotransmission. Neurotransmission at the NMJ is also highly modulated by muscarinic receptors M1 and M2 (mAChRs), which are related to PKA and TrkB signallings. Here, we investigated the hypothesis that TrkB, in cooperation with mAChRs, regulates the activity-dependent dynamics of PKA subunits to phosphorylate SNAP-25 and Synapsin-1. METHODS To explore this, we stimulated the rat phrenic nerve at 1Hz (30 minutes), with or without subsequent contraction (abolished by µ-conotoxin GIIIB). Pharmacological treatments were conducted with the anti-TrkB antibody clone 47/TrkB for TrkB inhibition and exogenous h-BDNF; muscarinic inhibition with Pirenzepine-dihydrochloride and Methoctramine-tetrahydrochloride for M1 and M2 mAChRs, respectively. Diaphragm protein levels and phosphorylation' changes were detected by Western blotting. Location of the target proteins was demonstrated using immunohistochemistry. RESULTS While TrkB does not directly impact the levels of PKA catalytic subunits Cα and Cβ, it regulates PKA regulatory subunits RIα and RIIβ, facilitating the phosphorylation of critical exocytotic targets such as SNAP-25 and Synapsin-1. Furthermore, the muscarinic receptors pathway maintains a delicate balance in this regulatory process. These findings explain the dynamic interplay of PKA subunits influenced by BDNF/TrkB signalling, M1 and M2 mAChRs pathways, that are differently regulated by pre- and postsynaptic activity, demonstrating the specific roles of the BDNF/TrkB and muscarinic receptors pathway in retrograde regulation. CONCLUSION This complex molecular interplay has the relevance of interrelating two fundamental pathways in PKA-synaptic modulation: one retrograde (neurotrophic) and the other autocrine (muscarinic). This deepens the fundamental understanding of neuromuscular physiology of neurotransmission that gives plasticity to synapses and holds the potential for identifying therapeutic strategies in conditions characterized by impaired neuromuscular communication.
Collapse
Affiliation(s)
- Aleksandra Polishchuk
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Víctor Cilleros-Mañé
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Marta Balanyà-Segura
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Laia Just-Borràs
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Anton Forniés-Mariné
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
| | - Carolina Silvera-Simón
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Marta Tomàs
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Meryem Jami El Hirchi
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Erica Hurtado
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Josep Tomàs
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Maria A Lanuza
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain.
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| |
Collapse
|
5
|
Hacisuleyman E, Hale CR, Noble N, Luo JD, Fak JJ, Saito M, Chen J, Weissman JS, Darnell RB. Neuronal activity rapidly reprograms dendritic translation via eIF4G2:uORF binding. Nat Neurosci 2024; 27:822-835. [PMID: 38589584 PMCID: PMC11088998 DOI: 10.1038/s41593-024-01615-5] [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: 08/03/2023] [Accepted: 03/05/2024] [Indexed: 04/10/2024]
Abstract
Learning and memory require activity-induced changes in dendritic translation, but which mRNAs are involved and how they are regulated are unclear. In this study, to monitor how depolarization impacts local dendritic biology, we employed a dendritically targeted proximity labeling approach followed by crosslinking immunoprecipitation, ribosome profiling and mass spectrometry. Depolarization of primary cortical neurons with KCl or the glutamate agonist DHPG caused rapid reprogramming of dendritic protein expression, where changes in dendritic mRNAs and proteins are weakly correlated. For a subset of pre-localized messages, depolarization increased the translation of upstream open reading frames (uORFs) and their downstream coding sequences, enabling localized production of proteins involved in long-term potentiation, cell signaling and energy metabolism. This activity-dependent translation was accompanied by the phosphorylation and recruitment of the non-canonical translation initiation factor eIF4G2, and the translated uORFs were sufficient to confer depolarization-induced, eIF4G2-dependent translational control. These studies uncovered an unanticipated mechanism by which activity-dependent uORF translational control by eIF4G2 couples activity to local dendritic remodeling.
Collapse
Affiliation(s)
- Ezgi Hacisuleyman
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA.
| | - Caryn R Hale
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natalie Noble
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller University, New York, NY, USA
| | - John J Fak
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Misa Saito
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Jin Chen
- Department of Pharmacology and Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Altos Labs, Bay Area Institute of Science, Redwood City, CA, USA
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Robert B Darnell
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
6
|
Ma S, Dou Y, Wang W, Wei A, Lan M, Liu J, Yang L, Yang M. Association between esketamine interventions and postpartum depression and analgesia following cesarean delivery: a systematic review and meta-analysis. Am J Obstet Gynecol MFM 2024; 6:101241. [PMID: 38262519 DOI: 10.1016/j.ajogmf.2023.101241] [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: 08/21/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/25/2024]
Abstract
OBJECTIVE This study aimed to compare the efficacy and safety of the use of esketamine to reduce the risk for postpartum depression and pain after cesarean delivery. DATA SOURCES Literature searches were conducted in PubMed, Embase, Cochrane Library, Web of Science, CNKI, and Wan fang from inception to August 2023. STUDY ELIGIBILITY CRITERIA The eligibility criteria were all randomized controlled trials of people who underwent a cesarean delivery and who were randomized to receive esketamine interventions irrespective of age or ethnicity. The outcomes that were assessed included the incidence of postpartum depression and the Edinburgh Postnatal Depression Scale score within 7 days and at 28 to 42 days after delivery, the pain score (visual analog scale or numerical rating scale, 0-10), the consumption of opioids, and intraoperative and postoperative adverse events. METHODS The Cochrane collaboration's tool was used for quality appraisal of the included studies. Statistical analysis of the data was performed using Review Manager 5.3 software, and the results were expressed as mean differences with 95% confidence intervals. Assessments were pooled using a random-effects or fixed-effects model. Study heterogeneity was assessed using the standard I2 statistic. RESULTS Among the 11 included randomized controlled trials that used the Edinburgh Postnatal Depression Scale for postpartum depression assessment, patients in esketamine group had a lower risk for postpartum depression within a week of surgery (risk ratio, 0.45; 95% confidence interval, 0.33-0.62). Intraoperative use of esketamine maintained a lower Edinburgh Postnatal Depression Scale score after surgery (mean difference, -1.64; 95% confidence interval, -2.14 to -1.14). Esketamine was associated with a beneficial effect in terms of the other outcomes, including a significant decline in pain score within 48 hours (mean difference, -0.71; 95% confidence interval, -0.89 to 0.52). Esketamine increased the risk for adverse neurologic and mental events during surgery without harming health, and there was no significant difference after delivery when compared with the control group. CONCLUSION Esketamine may reduce the risk for postpartum depression among patients who are undergoing cesarean delivery in the short term. In addition, as an adjunct to reduce analgesia, esketamine also effectively assists in pain management. Because of the lack of more high-quality evidence, we need more compelling evidence to confirm the value of esketamine in improving postpartum recovery.
Collapse
Affiliation(s)
- Shijin Ma
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (Ms Ma, Mr Dou, Ms Wang, Ms Wei, Ms Lan, Ms Liu, Ms Yang, and Dr Yang)
| | - Yuzhe Dou
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (Ms Ma, Mr Dou, Ms Wang, Ms Wei, Ms Lan, Ms Liu, Ms Yang, and Dr Yang)
| | - Wei Wang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (Ms Ma, Mr Dou, Ms Wang, Ms Wei, Ms Lan, Ms Liu, Ms Yang, and Dr Yang)
| | - Ai Wei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (Ms Ma, Mr Dou, Ms Wang, Ms Wei, Ms Lan, Ms Liu, Ms Yang, and Dr Yang)
| | - Mengxia Lan
- Chengdu University of Traditional Chinese Medicine, Chengdu, China (Ms Lan)
| | - Jingyu Liu
- Southwest Medical University, Luzhou, China (Ms Liu)
| | - Lina Yang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (Ms Ma, Mr Dou, Ms Wang, Ms Wei, Ms Lan, Ms Liu, Ms Yang, and Dr Yang).
| | - Mengchang Yang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (Ms Ma, Mr Dou, Ms Wang, Ms Wei, Ms Lan, Ms Liu, Ms Yang, and Dr Yang).
| |
Collapse
|
7
|
Johnson JL, Steele JH, Lin R, Stepanov VG, Gavriliuc MN, Wang Y. Multi-Channel smFRET study reveals a Compact conformation of EF-G on the Ribosome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.577133. [PMID: 38328191 PMCID: PMC10849647 DOI: 10.1101/2024.01.27.577133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
While elongation factor G (EF-G) is crucial for ribosome translocation, the role of its GTP hydrolysis remains ambiguous. EF-G's indispensability is further exemplified by the phosphorylation of human eukaryotic elongation factor 2 (eEF2) at Thr56, which inhibits protein synthesis globally, but its exact mechanism is not clear. In this study, we developed a multi-channel single-molecule FRET (smFRET) microscopy methodology to examine the conformational changes of E. coli EF-G induced by mutations that closely aligned with eEF2's Thr56 residue. We utilized Alexa 488/594 double-labeled EF-G to catalyze the translocation of fMet-Phe-tRNAPhe-Cy3 inside Cy5-L27 labeled ribosomes, allowing us to probe both processes within the same complex. Our findings indicate that in the presence of either GTP or GDPCP, wild-type EF-G undergoes a conformational extension upon binding to the ribosome to promote normal translocation. On the other hand, T48E and T48V mutations did not affect GTP/GDP binding or GTP hydrolysis, but impeded Poly(Phe) synthesis and caused EF-G to adopt a unique compact conformation, which wasn't observed when the mutants interact solely with the sarcin/ricin loop. This study provides new insights into EF-G's adaptability and sheds light on the modification mechanism of human eEF2.
Collapse
Affiliation(s)
- Jordan L Johnson
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Jacob H Steele
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Ran Lin
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Victor G Stepanov
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Miriam N Gavriliuc
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Yuhong Wang
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
8
|
Fernandez A, Corvalan K, Santis O, Mendez-Ruette M, Caviedes A, Pizarro M, Gomez MT, Batiz LF, Landgraf P, Kahne T, Rojas-Fernandez A, Wyneken U. Sumoylation in astrocytes induces changes in the proteome of the derived small extracellular vesicles which change protein synthesis and dendrite morphology in target neurons. Brain Res 2024; 1823:148679. [PMID: 37972846 DOI: 10.1016/j.brainres.2023.148679] [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: 08/25/2023] [Revised: 11/01/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Emerging evidence highlights the relevance of the protein post-translational modification by SUMO (Small Ubiquitin-like Modifier) in the central nervous system for modulating cognition and plasticity in health and disease. In these processes, astrocyte-to-neuron crosstalk mediated by extracellular vesicles (EVs) plays a yet poorly understood role. Small EVs (sEVs), including microvesicles and exosomes, contain a molecular cargo of lipids, proteins, and nucleic acids that define their biological effect on target cells. Here, we investigated whether SUMOylation globally impacts the sEV protein cargo. For this, sEVs were isolated from primary cultures of astrocytes by ultracentrifugation or using a commercial sEV isolation kit. SUMO levels were regulated: 1) via plasmids that over-express SUMO, or 2) via experimental conditions that increase SUMOylation, i.e., by using the stress hormone corticosterone, or 3) via the SUMOylation inhibitor 2-D08 (2',3',4'-trihydroxy-flavone, 2-(2,3,4-Trihydroxyphenyl)-4H-1-Benzopyran-4-one). Corticosterone and 2-D08 had opposing effects on the number of sEVs and on their protein cargo. Proteomic analysis showed that increased SUMOylation in corticosterone-treated or plasmid-transfected astrocytes increased the presence of proteins related to cell division, transcription, and protein translation in the derived sEVs. When sEVs derived from corticosterone-treated astrocytes were transferred to neurons to assess their impact on protein synthesis using the fluorescence non-canonical amino acid tagging assay (FUNCAT), we detected an increase in protein synthesis, while sEVs from 2-D08-treated astrocytes had no effect. Our results show that SUMO conjugation plays an important role in the modulation of the proteome of astrocyte-derived sEVs with a potential functional impact on neurons.
Collapse
Affiliation(s)
- Anllely Fernandez
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Katherine Corvalan
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Octavia Santis
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Maxs Mendez-Ruette
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Ariel Caviedes
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Matias Pizarro
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Maria-Teresa Gomez
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Luis Federico Batiz
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Peter Landgraf
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Germany, 39120 Magdeburg, Germany
| | - Thilo Kahne
- Institute of Experimental Internal Medicine, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Alejandro Rojas-Fernandez
- Instituto de Medicina & Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Ursula Wyneken
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7620001, Chile.
| |
Collapse
|
9
|
Vilchinskaya N, Lim WF, Belova S, Roberts TC, Wood MJA, Lomonosova Y. Investigating Eukaryotic Elongation Factor 2 Kinase/Eukaryotic Translation Elongation Factor 2 Pathway Regulation and Its Role in Protein Synthesis Impairment during Disuse-Induced Skeletal Muscle Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2023:S0002-9440(23)00060-3. [PMID: 36871751 DOI: 10.1016/j.ajpath.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/16/2023] [Accepted: 02/10/2023] [Indexed: 03/07/2023]
Abstract
The principal mechanism underlying the reduced rate of protein synthesis in atrophied skeletal muscle is largely unknown. Eukaryotic elongation factor 2 kinase (eEF2k) impairs the ability of eukaryotic translation elongation factor 2 (eEF2) to bind to the ribosome via T56 phosphorylation. Perturbations in the eEF2k/eEF2 pathway during various stages of disuse muscle atrophy have been investigated utilizing a rat hind limb suspension (HS) model. Two distinct components of eEF2k/eEF2 pathway misregulation were demonstrated, observing a significant (P < 0.01) increase in eEF2k mRNA expression as early as 1-day HS and in eEF2k protein level after 3-day HS. We set out to determine whether eEF2k activation is a Ca2+-dependent process with involvement of Cav1.1. The ratio of T56-phosphorylated/total eEF2 was robustly elevated after 3-day HS, which was completely reversed by BAPTA-AM and decreased by 1.7-fold (P < 0.05) by nifedipine. Transfection of C2C12 with pCMV-eEF2k and administration with small molecules were used to modulate eEF2k and eEF2 activity. More important, pharmacologic enhancement of eEF2 phosphorylation induced phosphorylated ribosomal protein S6 kinase (T389) up-regulation and restoration of global protein synthesis in the HS rats. Taken together, the eEF2k/eEF2 pathway is up-regulated during disuse muscle atrophy involving calcium-dependent activation of eEF2k partly via Cav1.1. The study provides evidence, in vitro and in vivo, of the eEF2k/eEF2 pathway impact on ribosomal protein S6 kinase activity as well as protein expression of key atrophy biomarkers, muscle atrophy F-box/atrogin-1 and muscle RING finger-1.
Collapse
Affiliation(s)
| | - Wooi Fang Lim
- Department of Paediatrics, University of Oxford Children's Hospital, John Radcliffe Hospital, Oxford, United Kingdom; Institute of Developmental and Regenerative Medicine, Oxford, United Kingdom; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom
| | | | - Thomas C Roberts
- Department of Paediatrics, University of Oxford Children's Hospital, John Radcliffe Hospital, Oxford, United Kingdom; Institute of Developmental and Regenerative Medicine, Oxford, United Kingdom; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford Children's Hospital, John Radcliffe Hospital, Oxford, United Kingdom; Institute of Developmental and Regenerative Medicine, Oxford, United Kingdom; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom
| | - Yulia Lomonosova
- Department of Paediatrics, University of Oxford Children's Hospital, John Radcliffe Hospital, Oxford, United Kingdom; Institute of Developmental and Regenerative Medicine, Oxford, United Kingdom; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
10
|
Polishchuk A, Cilleros-Mañé V, Just-Borràs L, Balanyà-Segura M, Vandellòs Pont G, Silvera Simón C, Tomàs M, Garcia N, Tomàs J, Lanuza MA. Synaptic retrograde regulation of the PKA-induced SNAP-25 and Synapsin-1 phosphorylation. Cell Mol Biol Lett 2023; 28:17. [PMID: 36869288 PMCID: PMC9985302 DOI: 10.1186/s11658-023-00431-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/09/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Bidirectional communication between presynaptic and postsynaptic components contribute to the homeostasis of the synapse. In the neuromuscular synapse, the arrival of the nerve impulse at the presynaptic terminal triggers the molecular mechanisms associated with ACh release, which can be retrogradely regulated by the resulting muscle contraction. This retrograde regulation, however, has been poorly studied. At the neuromuscular junction (NMJ), protein kinase A (PKA) enhances neurotransmitter release, and the phosphorylation of the molecules of the release machinery including synaptosomal associated protein of 25 kDa (SNAP-25) and Synapsin-1 could be involved. METHODS Accordingly, to study the effect of synaptic retrograde regulation of the PKA subunits and its activity, we stimulated the rat phrenic nerve (1 Hz, 30 min) resulting or not in contraction (abolished by µ-conotoxin GIIIB). Changes in protein levels and phosphorylation were detected by western blotting and cytosol/membrane translocation by subcellular fractionation. Synapsin-1 was localized in the levator auris longus (LAL) muscle by immunohistochemistry. RESULTS Here we show that synaptic PKA Cβ subunit regulated by RIIβ or RIIα subunits controls activity-dependent phosphorylation of SNAP-25 and Synapsin-1, respectively. Muscle contraction retrogradely downregulates presynaptic activity-induced pSynapsin-1 S9 while that enhances pSNAP-25 T138. Both actions could coordinately contribute to decreasing the neurotransmitter release at the NMJ. CONCLUSION This provides a molecular mechanism of the bidirectional communication between nerve terminals and muscle cells to balance the accurate process of ACh release, which could be important to characterize molecules as a therapy for neuromuscular diseases in which neuromuscular crosstalk is impaired.
Collapse
Affiliation(s)
- Aleksandra Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Balanyà-Segura
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Genís Vandellòs Pont
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Carolina Silvera Simón
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain.
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
11
|
Ma X, Li L, Li Z, Huang Z, Yang Y, Liu P, Guo D, Li Y, Wu T, Luo R, Xu J, Ye W, Jiang B, Shi L. eEF2 in the prefrontal cortex promotes excitatory synaptic transmission and social novelty behavior. EMBO Rep 2022; 23:e54543. [PMID: 35993189 PMCID: PMC9535807 DOI: 10.15252/embr.202154543] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/22/2022] [Accepted: 08/03/2022] [Indexed: 08/24/2023] Open
Abstract
Regulation of mRNA translation is essential for brain development and function. Translation elongation factor eEF2 acts as a molecular hub orchestrating various synaptic signals to protein synthesis control and participates in hippocampus-dependent cognitive functions. However, whether eEF2 regulates other behaviors in different brain regions has been unknown. Here, we construct a line of Eef2 heterozygous (HET) mice, which show a reduction in eEF2 and protein synthesis mainly in excitatory neurons of the prefrontal cortex. The mice also show lower spine density, reduced excitability, and AMPAR-mediated synaptic transmission in pyramidal neurons of the medial prefrontal cortex (mPFC). While HET mice exhibit normal learning and memory, they show defective social behavior and elevated anxiety. Knockdown of Eef2 in excitatory neurons of the mPFC specifically is sufficient to impair social novelty preference. Either chemogenetic activation of excitatory neurons in the mPFC or mPFC local infusion of the AMPAR potentiator PF-4778574 corrects the social novelty deficit of HET mice. Collectively, we identify a novel role for eEF2 in promoting prefrontal AMPAR-mediated synaptic transmission underlying social novelty behavior.
Collapse
Affiliation(s)
- Xuanyue Ma
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Liuren Li
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Ziming Li
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Zhengyi Huang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Yaorong Yang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Peng Liu
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Daji Guo
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
- Clinical Neuroscience InstituteThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yueyao Li
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Tianying Wu
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Ruixiang Luo
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Junyu Xu
- Department of Neurobiology and Department of Rehabilitation of the Children's HospitalZhejiang University School of MedicineHangzhouChina
| | - Wen‐Cai Ye
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
- Center for Bioactive Natural Molecules and Innovative Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Bin Jiang
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Lei Shi
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
- Center for Bioactive Natural Molecules and Innovative Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of PharmacyJinan UniversityGuangzhouChina
| |
Collapse
|
12
|
Zhu Z, Fang C, Xu H, Yuan L, Du Y, Ni Y, Xu Y, Shao A, Zhang A, Lou M. Anoikis resistance in diffuse glioma: The potential therapeutic targets in the future. Front Oncol 2022; 12:976557. [PMID: 36046036 PMCID: PMC9423707 DOI: 10.3389/fonc.2022.976557] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Glioma is the most common malignant intracranial tumor and exhibits diffuse metastasis and a high recurrence rate. The invasive property of glioma results from cell detachment. Anoikis is a special form of apoptosis that is activated upon cell detachment. Resistance to anoikis has proven to be a protumor factor. Therefore, it is suggested that anoikis resistance commonly occurs in glioma and promotes diffuse invasion. Several factors, such as integrin, E-cadherin, EGFR, IGFR, Trk, TGF-β, the Hippo pathway, NF-κB, eEF-2 kinase, MOB2, hypoxia, acidosis, ROS, Hsp and protective autophagy, have been shown to induce anoikis resistance in glioma. In our present review, we aim to summarize the underlying mechanism of resistance and the therapeutic potential of these molecules.
Collapse
Affiliation(s)
- Zhengyang Zhu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichao Du
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunjia Ni
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanzhi Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Anke Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Tang F, Fan J, Zhang X, Zou Z, Xiao D, Li X. The Role of Vti1a in Biological Functions and Its Possible Role in Nervous System Disorders. Front Mol Neurosci 2022; 15:918664. [PMID: 35711736 PMCID: PMC9197314 DOI: 10.3389/fnmol.2022.918664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022] Open
Abstract
Vesicle transport through interaction with t-SNAREs 1A (Vti1a), a member of the N-ethylmaleimide-sensitive factor attachment protein receptor protein family, is involved in cell signaling as a vesicular protein and mediates vesicle trafficking. Vti1a appears to have specific roles in neurons, primarily by regulating upstream neurosecretory events that mediate exocytotic proteins and the availability of secretory organelles, as well as regulating spontaneous synaptic transmission and postsynaptic efficacy to control neurosecretion. Vti1a also has essential roles in neural development, autophagy, and unconventional extracellular transport of neurons. Studies have shown that Vti1a dysfunction plays critical roles in pathological mechanisms of Hepatic encephalopathy by influencing spontaneous neurotransmission. It also may have an unknown role in amyotrophic lateral sclerosis. A VTI1A variant is associated with the risk of glioma, and the fusion product of the VTI1A gene and the adjacent TCF7L2 gene is involved in glioma development. This review summarizes Vti1a functions in neurons and highlights the role of Vti1a in the several nervous system disorders.
Collapse
Affiliation(s)
- Fajuan Tang
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Jiali Fan
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaoyan Zhang
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Zhuan Zou
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Dongqiong Xiao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Dongqiong Xiao,
| | - Xihong Li
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- *Correspondence: Xihong Li,
| |
Collapse
|
14
|
Abstract
N-methyl-d-aspartate receptors (NMDARs) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) are excitatory neurotransmission receptors of the central nervous system and play vital roles in synaptic plasticity. Although not fully elucidated, visceral hypersensitivity is one of the most well-characterized pathophysiologic abnormalities of functional gastrointestinal diseases and appears to be associated with increased synaptic plasticity. In this study, we review the updated findings on the physiology of NMDARs and AMPARs and their relation to visceral hypersensitivity, which propose directions for future research in this field with evolving importance.
Collapse
|
15
|
A single intravenous administration of a sub-anesthetic ketamine dose during the perioperative period of cesarean section for preventing postpartum depression: A meta-analysis. Psychiatry Res 2022; 310:114396. [PMID: 35278826 DOI: 10.1016/j.psychres.2022.114396] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/23/2022]
Abstract
The feasibility of intravenous ketamine administration during the perioperative period of cesarean section to prevent postpartum depression (PPD) has not been determined by meta-analysis. To evaluate the efficacy, safety and dose of prophylactic ketamine in offsetting PPD, we retrieved the following databases in English or Chinese from inception to December 2020: Pubmed, Embase, Web of Science, The Cochrane Library, CNKI, VIP and Wanfang. A total of 10 studies (9 RCTs and 1 retrospective study) were included with 2087 cases. Meta-analysis showed that in ketamine group, the score and the prevalence of PPD within 1 week postpartum were significantly reduced, whereas PPD score after 4 weeks postpartum showed no superiority. There was no significant difference in terms of total adverse events rate, although vomiting occurred more frequently in the ketamine group. In addition, we found that ketamine efficacy emerged at 0.5 mg/kg. By meta-regression, we observed that: (1) Age and BMI are negatively associated with mood response to ketamine. (2) An analgesic pump containing ketamine for continuous 48 h postpartum administration was more efficacious than an intravenous injection of ketamine during cesarean section. Current evidence shows ketamine could be efficacious and safe in the prophylactic management of PPD in women having a cesarean section.
Collapse
|
16
|
Beretta S, Gritti L, Ponzoni L, Scalmani P, Mantegazza M, Sala M, Verpelli C, Sala C. Rescuing epileptic and behavioral alterations in a Dravet syndrome mouse model by inhibiting eukaryotic elongation factor 2 kinase (eEF2K). Mol Autism 2022; 13:1. [PMID: 34980259 PMCID: PMC8722032 DOI: 10.1186/s13229-021-00484-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022] Open
Abstract
Background Dravet Syndrome is a severe childhood pharmaco-resistant epileptic disorder mainly caused by mutations in the SCN1A gene, which encodes for the α1 subunit of the type I voltage-gated sodium channel (NaV1.1), that causes imbalance between excitation and inhibition in the brain. We recently found that eEF2K knock out mice displayed enhanced GABAergic transmission and tonic inhibition and were less susceptible to epileptic seizures. Thus, we investigated the effect of inhibition of eEF2K on the epileptic and behavioral phenotype of Scn1a ± mice, a murine model of Dravet Syndrome. Methods To elucidate the role of eEF2K pathway in the etiopathology of Dravet syndrome we generated a new mouse model deleting the eEF2K gene in Scn1a ± mice. By crossing Scn1a ± mice with eEF2K−/− mice we obtained the three main genotypes needed for our studies, Scn1a+/+ eEF2K+/+ (WT mice), Scn1a ± eEF2K+/+ mice (Scn1a ± mice) and Scn1a ± eEF2K−/− mice, that were fully characterized for EEG and behavioral phenotype. Furthermore, we tested the ability of a pharmacological inhibitor of eEF2K in rescuing EEG alterations of the Scn1a ± mice. Results We showed that the activity of eEF2K/eEF2 pathway was enhanced in Scn1a ± mice. Then, we demonstrated that both genetic deletion and pharmacological inhibition of eEF2K were sufficient to ameliorate the epileptic phenotype of Scn1a ± mice. Interestingly we also found that motor coordination defect, memory impairments, and stereotyped behavior of the Scn1a ± mice were reverted by eEF2K deletion. The analysis of spontaneous inhibitory postsynaptic currents (sIPSCs) suggested that the rescue of the pathological phenotype was driven by the potentiation of GABAergic synapses. Limitations Even if we found that eEF2K deletion was able to increase inhibitory synapses function, the molecular mechanism underlining the inhibition of eEF2K/eEF2 pathway in rescuing epileptic and behavioral alterations in the Scn1a ± needs further investigations. Conclusions Our data indicate that pharmacological inhibition of eEF2K could represent a novel therapeutic intervention for treating epilepsy and related comorbidities in the Dravet syndrome. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00484-0.
Collapse
Affiliation(s)
- Stefania Beretta
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Laura Gritti
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Luisa Ponzoni
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Paolo Scalmani
- L'Unità Operativa Complessa di Epilettologia Clinica e Sperimentale, Foundation Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Neurological Institute Carlo Besta, 20133, Milan, Italy
| | - Massimo Mantegazza
- CNRS UMR 7275, Institut National de La Santé Et de La Recherche Médicale, LabEx ICST, Institute of Molecular and Cellular Pharmacology (IPMC), Université Côte d'Azur (UCA), 06560, Valbonne-Sophia Antipolis, France
| | - Mariaelvina Sala
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| | - Carlo Sala
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| |
Collapse
|
17
|
Smith PR, Loerch S, Kunder N, Stanowick AD, Lou TF, Campbell ZT. Functionally distinct roles for eEF2K in the control of ribosome availability and p-body abundance. Nat Commun 2021; 12:6789. [PMID: 34815424 PMCID: PMC8611098 DOI: 10.1038/s41467-021-27160-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/07/2021] [Indexed: 11/09/2022] Open
Abstract
Processing bodies (p-bodies) are a prototypical phase-separated RNA-containing granule. Their abundance is highly dynamic and has been linked to translation. Yet, the molecular mechanisms responsible for coordinate control of the two processes are unclear. Here, we uncover key roles for eEF2 kinase (eEF2K) in the control of ribosome availability and p-body abundance. eEF2K acts on a sole known substrate, eEF2, to inhibit translation. We find that the eEF2K agonist nelfinavir abolishes p-bodies in sensory neurons and impairs translation. To probe the latter, we used cryo-electron microscopy. Nelfinavir stabilizes vacant 80S ribosomes. They contain SERBP1 in place of mRNA and eEF2 in the acceptor site. Phosphorylated eEF2 associates with inactive ribosomes that resist splitting in vitro. Collectively, the data suggest that eEF2K defines a population of inactive ribosomes resistant to recycling and protected from degradation. Thus, eEF2K activity is central to both p-body abundance and ribosome availability in sensory neurons.
Collapse
Affiliation(s)
- Patrick R. Smith
- grid.267323.10000 0001 2151 7939The University of Texas at Dallas, Department of Biological Sciences, Richardson, TX USA
| | - Sarah Loerch
- grid.443970.dJanelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA USA ,grid.205975.c0000 0001 0740 6917University of California, Santa Cruz, Department of Chemistry and Biochemistry, Santa Cruz, CA USA
| | - Nikesh Kunder
- grid.267323.10000 0001 2151 7939The University of Texas at Dallas, Department of Biological Sciences, Richardson, TX USA
| | - Alexander D. Stanowick
- grid.267323.10000 0001 2151 7939The University of Texas at Dallas, Department of Biological Sciences, Richardson, TX USA
| | - Tzu-Fang Lou
- grid.267323.10000 0001 2151 7939The University of Texas at Dallas, Department of Biological Sciences, Richardson, TX USA
| | - Zachary T. Campbell
- grid.267323.10000 0001 2151 7939The University of Texas at Dallas, Department of Biological Sciences, Richardson, TX USA ,grid.267323.10000 0001 2151 7939The Center for Advanced Pain Studies (CAPS), University of Texas at Dallas, Richardson, TX USA
| |
Collapse
|
18
|
Kowalczyk M, Kowalczyk E, Kwiatkowski P, Łopusiewicz Ł, Sienkiewicz M, Talarowska M. Ketamine-New Possibilities in the Treatment of Depression: A Narrative Review. Life (Basel) 2021; 11:1186. [PMID: 34833062 PMCID: PMC8619908 DOI: 10.3390/life11111186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 12/11/2022] Open
Abstract
The SARS-CoV-2 coronavirus epidemic has led to an increase in the number of people with depression. Symptoms related to the mental sphere (mainly depression and anxiety) may be experienced by one third of the worldwide population. This entails the need for the effective and rapid treatment of depressive episodes. An effective drug seems to be s-ketamine, which was accepted in March 2019 by the Food and Drug Administration (FDA) for the treatment of drug-resistant depression. This drug provides a quick antidepressant effect with maximum effectiveness achieved after 24 h. It also appears to reduce the occurrence of suicidal thoughts. However, research into undesirable effects, especially in groups of people susceptible to psychotic episodes or those who use alcohol or psychoactive substances, is necessary.
Collapse
Affiliation(s)
- Mateusz Kowalczyk
- Babinski Memorial Hospital, Aleksandrowska St. 159, 91-229 Lodz, Poland;
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego St. 7/9, 90-752 Lodz, Poland;
| | - Paweł Kwiatkowski
- Department of Diagnostic Immunology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Łukasz Łopusiewicz
- Center of Bioimmobilisation and Innovative Packaging Materials, Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Janickiego 35, 71-270 Szczecin, Poland;
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Muszyńskiego St. 1, 90-151 Lodz, Poland
| | - Monika Talarowska
- Department of Clinical Psychology and Psychopathology, Institute of Psychology, University of Lodz, Smugowa St. 10/12, 91-433 Lodz, Poland;
| |
Collapse
|
19
|
APOE4 Affects Basal and NMDAR-Mediated Protein Synthesis in Neurons by Perturbing Calcium Homeostasis. J Neurosci 2021; 41:8686-8709. [PMID: 34475200 DOI: 10.1523/jneurosci.0435-21.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/15/2021] [Accepted: 08/22/2021] [Indexed: 01/24/2023] Open
Abstract
Apolipoprotein E (APOE), one of the primary lipoproteins in the brain has three isoforms in humans, APOE2, APOE3, and APOE4. APOE4 is the most well-established risk factor increasing the predisposition for Alzheimer's disease (AD). The presence of the APOE4 allele alone is shown to cause synaptic defects in neurons and recent studies have identified multiple pathways directly influenced by APOE4. However, the mechanisms underlying APOE4-induced synaptic dysfunction remain elusive. Here, we report that the acute exposure of primary cortical neurons or synaptoneurosomes to APOE4 leads to a significant decrease in global protein synthesis. Primary cortical neurons were derived from male and female embryos of Sprague Dawley (SD) rats or C57BL/6J mice. Synaptoneurosomes were prepared from P30 male SD rats. APOE4 treatment also abrogates the NMDA-mediated translation response indicating an alteration of synaptic signaling. Importantly, we demonstrate that both APOE3 and APOE4 generate a distinct translation response which is closely linked to their respective calcium signature. Acute exposure of neurons to APOE3 causes a short burst of calcium through NMDA receptors (NMDARs) leading to an initial decrease in protein synthesis which quickly recovers. Contrarily, APOE4 leads to a sustained increase in calcium levels by activating both NMDARs and L-type voltage-gated calcium channels (L-VGCCs), thereby causing sustained translation inhibition through eukaryotic translation elongation factor 2 (eEF2) phosphorylation, which in turn disrupts the NMDAR response. Thus, we show that APOE4 affects basal and activity-mediated protein synthesis responses in neurons by affecting calcium homeostasis.SIGNIFICANCE STATEMENT Defective protein synthesis has been shown as an early defect in familial Alzheimer's disease (AD). However, this has not been studied in the context of sporadic AD, which constitutes the majority of cases. In our study, we show that Apolipoprotein E4 (APOE4), the predominant risk factor for AD, inhibits global protein synthesis in neurons. APOE4 also affects NMDA activity-mediated protein synthesis response, thus inhibiting synaptic translation. We also show that the defective protein synthesis mediated by APOE4 is closely linked to the perturbation of calcium homeostasis caused by APOE4 in neurons. Thus, we propose the dysregulation of protein synthesis as one of the possible molecular mechanisms to explain APOE4-mediated synaptic and cognitive defects. Hence, the study not only suggests an explanation for the APOE4-mediated predisposition to AD, it also bridges the gap in understanding APOE4-mediated pathology.
Collapse
|
20
|
Ballard DJ, Peng HY, Das JK, Kumar A, Wang L, Ren Y, Xiong X, Ren X, Yang JM, Song J. Insights Into the Pathologic Roles and Regulation of Eukaryotic Elongation Factor-2 Kinase. Front Mol Biosci 2021; 8:727863. [PMID: 34532346 PMCID: PMC8438118 DOI: 10.3389/fmolb.2021.727863] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic Elongation Factor-2 Kinase (eEF2K) acts as a negative regulator of protein synthesis, translation, and cell growth. As a structurally unique member of the alpha-kinase family, eEF2K is essential to cell survival under stressful conditions, as it contributes to both cell viability and proliferation. Known as the modulator of the global rate of protein translation, eEF2K inhibits eEF2 (eukaryotic Elongation Factor 2) and decreases translation elongation when active. eEF2K is regulated by various mechanisms, including phosphorylation through residues and autophosphorylation. Specifically, this protein kinase is downregulated through the phosphorylation of multiple sites via mTOR signaling and upregulated via the AMPK pathway. eEF2K plays important roles in numerous biological systems, including neurology, cardiology, myology, and immunology. This review provides further insights into the current roles of eEF2K and its potential to be explored as a therapeutic target for drug development.
Collapse
Affiliation(s)
- Darby J. Ballard
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Hao-Yun Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Anil Kumar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Yijie Ren
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
21
|
Cilleros-Mañé V, Just-Borràs L, Polishchuk A, Durán M, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. M 1 and M 2 mAChRs activate PDK1 and regulate PKC βI and ε and the exocytotic apparatus at the NMJ. FASEB J 2021; 35:e21724. [PMID: 34133802 DOI: 10.1096/fj.202002213r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Neuromuscular junctions (NMJ) regulate cholinergic exocytosis through the M1 and M2 muscarinic acetylcholine autoreceptors (mAChR), involving the crosstalk between receptors and downstream pathways. Protein kinase C (PKC) regulates neurotransmission but how it associates with the mAChRs remains unknown. Here, we investigate whether mAChRs recruit the classical PKCβI and the novel PKCε isoforms and modulate their priming by PDK1, translocation and activity on neurosecretion targets. We show that each M1 and M2 mAChR activates the master kinase PDK1 and promotes a particular priming of the presynaptic PKCβI and ε isoforms. M1 recruits both primed-PKCs to the membrane and promotes Munc18-1, SNAP-25, and MARCKS phosphorylation. In contrast, M2 downregulates PKCε through a PKA-dependent pathway, which inhibits Munc18-1 synthesis and PKC phosphorylation. In summary, our results discover a co-dependent balance between muscarinic autoreceptors which orchestrates the presynaptic PKC and their action on ACh release SNARE-SM mechanism. Altogether, this molecular signaling explains previous functional studies at the NMJ and guide toward potential therapeutic targets.
Collapse
Affiliation(s)
- V Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - L Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - A Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Durán
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - N Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - J M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
22
|
Shen Y, Han J, Zhang ZC. Novel regulation of the eEF2K/eEF2 pathway: prospects of 'PQBP1 promotes translational elongation and regulates hippocampal mGluR-LTD by suppressing eEF2 phosphorylation'. J Mol Cell Biol 2021; 13:392-394. [PMID: 33734395 PMCID: PMC8373267 DOI: 10.1093/jmcb/mjab017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Yuqian Shen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junhai Han
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zi Chao Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
23
|
Mitroshina EV, Mishchenko TA, Loginova MM, Tarabykin VS, Vedunova MV. Identification of Kinome Representatives with Neuroprotective Activity. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420040133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
24
|
Rana T, Behl T, Sehgal A, Srivastava P, Bungau S. Unfolding the Role of BDNF as a Biomarker for Treatment of Depression. J Mol Neurosci 2020; 71:2008-2021. [PMID: 33230708 DOI: 10.1007/s12031-020-01754-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/08/2020] [Indexed: 12/13/2022]
Abstract
Depression is a well-known disabling mental illness characterized by sadness, loss of interest in activities, and decreased energy. The symptoms of depression are usually recurrent in vulnerable individuals, and persistence of symptoms significantly impairs individuals' quality of life. The exact pathophysiology of depression remains ambiguous, though many hypotheses have been proposed. Brain-derived neurotrophic factor (BDNF) has recently been reported to play a vital role in the pathophysiology of depression. BDNF is an important neurotrophic factor found in the human brain and is involved in neuronal growth and proliferation, synaptic neurotransmission, and neuroplasticity. The neurotrophic theory of depression proposes that depression results from reduced BDNF levels in the brain, which can be treated with antidepressants to alleviate depressive behavior and increase BDNF levels. The aim of this review is to provide broad insight into the role of BDNF in the pathogenesis of depression and in antidepressant therapy. The studies mentioned in this review article greatly support the role of BDNF in the pathogenesis of depression and treatment of this disorder with antidepressants. Since abnormalities in BDNF levels lead to the production of diverse insults that amplify the development or progression of depression, it is important to study and explore BDNF impairment in relation to depression, neuroplasticity, and neurogenesis, and increasing BDNF levels through antidepressant therapy, showing positive response in the management of depression.
Collapse
Affiliation(s)
- Tarapati Rana
- Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | | | - Simona Bungau
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
25
|
Gladulich LFH, Peixoto-Rodrigues MC, Campello-Costa P, Paes-de-Carvalho R, Cossenza M. NMDA-induced nitric oxide generation and CREB activation in central nervous system is dependent on eukaryotic elongation factor 2 kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118783. [DOI: 10.1016/j.bbamcr.2020.118783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 01/28/2023]
|
26
|
Gladulich LFH, Xie J, Jensen KB, Kamei M, Paes-de-Carvalho R, Cossenza M, Proud CG. Bicuculline regulated protein synthesis is dependent on Homer1 and promotes its interaction with eEF2K through mTORC1-dependent phosphorylation. J Neurochem 2020; 157:1086-1101. [PMID: 32892352 DOI: 10.1111/jnc.15178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/29/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
The regulation of protein synthesis is a vital and finely tuned process in cellular physiology. In neurons, this process is very precisely regulated, as which mRNAs undergo translation is highly dependent on context. One of the most prominent regulators of protein synthesis is the enzyme eukaryotic elongation factor kinase 2 (eEF2K) that regulates the elongation stage of protein synthesis. This kinase and its substrate, eukaryotic elongation factor 2 (eEF2) are important in processes such as neuronal development and synaptic plasticity. eEF2K is regulated by multiple mechanisms including Ca2+ -ions and the mTORC1 signaling pathway, both of which play key roles in neurological processes such as learning and memory. In such settings, the localized control of protein synthesis is of crucial importance. In this work, we sought to investigate how the localization of eEF2K is controlled and the impact of this on protein synthesis in neuronal cells. In this study, we used both SH-SY5Y neuroblastoma cells and mouse cortical neurons, and pharmacologically and/or genetic approaches to modify eEF2K function. We show that eEF2K activity and localization can be regulated by its binding partner Homer1b/c, a scaffolding protein known for its participation in calcium-regulated signaling pathways. Furthermore, our results indicate that this interaction is regulated by the mTORC1 pathway, through a known phosphorylation site in eEF2K (S396), and that it affects rates of localized protein synthesis at synapses depending on the presence or absence of this scaffolding protein.
Collapse
Affiliation(s)
- Luis F H Gladulich
- Program of Neurosciences, Fluminense Federal University, Niterói, Brazil.,Lifelong Health, South Australia Health & Medical Research Institute (SAHMRI) Adelaide, SA, Australia
| | - Jianling Xie
- Lifelong Health, South Australia Health & Medical Research Institute (SAHMRI) Adelaide, SA, Australia
| | - Kirk B Jensen
- Lifelong Health, South Australia Health & Medical Research Institute (SAHMRI) Adelaide, SA, Australia
| | - Makoto Kamei
- Lifelong Health, South Australia Health & Medical Research Institute (SAHMRI) Adelaide, SA, Australia.,Center for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Roberto Paes-de-Carvalho
- Program of Neurosciences, Fluminense Federal University, Niterói, Brazil.,Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Marcelo Cossenza
- Program of Neurosciences, Fluminense Federal University, Niterói, Brazil.,Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - Christopher G Proud
- Lifelong Health, South Australia Health & Medical Research Institute (SAHMRI) Adelaide, SA, Australia
| |
Collapse
|
27
|
Lottes EN, Cox DN. Homeostatic Roles of the Proteostasis Network in Dendrites. Front Cell Neurosci 2020; 14:264. [PMID: 33013325 PMCID: PMC7461941 DOI: 10.3389/fncel.2020.00264] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Cellular protein homeostasis, or proteostasis, is indispensable to the survival and function of all cells. Distinct from other cell types, neurons are long-lived, exhibiting architecturally complex and diverse multipolar projection morphologies that can span great distances. These properties present unique demands on proteostatic machinery to dynamically regulate the neuronal proteome in both space and time. Proteostasis is regulated by a distributed network of cellular processes, the proteostasis network (PN), which ensures precise control of protein synthesis, native conformational folding and maintenance, and protein turnover and degradation, collectively safeguarding proteome integrity both under homeostatic conditions and in the contexts of cellular stress, aging, and disease. Dendrites are equipped with distributed cellular machinery for protein synthesis and turnover, including dendritically trafficked ribosomes, chaperones, and autophagosomes. The PN can be subdivided into an adaptive network of three major functional pathways that synergistically govern protein quality control through the action of (1) protein synthesis machinery; (2) maintenance mechanisms including molecular chaperones involved in protein folding; and (3) degradative pathways (e.g., Ubiquitin-Proteasome System (UPS), endolysosomal pathway, and autophagy. Perturbations in any of the three arms of proteostasis can have dramatic effects on neurons, especially on their dendrites, which require tightly controlled homeostasis for proper development and maintenance. Moreover, the critical importance of the PN as a cell surveillance system against protein dyshomeostasis has been highlighted by extensive work demonstrating that the aggregation and/or failure to clear aggregated proteins figures centrally in many neurological disorders. While these studies demonstrate the relevance of derangements in proteostasis to human neurological disease, here we mainly review recent literature on homeostatic developmental roles the PN machinery plays in the establishment, maintenance, and plasticity of stable and dynamic dendritic arbors. Beyond basic housekeeping functions, we consider roles of PN machinery in protein quality control mechanisms linked to dendritic plasticity (e.g., dendritic spine remodeling during LTP); cell-type specificity; dendritic morphogenesis; and dendritic pruning.
Collapse
Affiliation(s)
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
28
|
David O, Barrera I, Gould N, Gal-Ben-Ari S, Rosenblum K. D1 Dopamine Receptor Activation Induces Neuronal eEF2 Pathway-Dependent Protein Synthesis. Front Mol Neurosci 2020; 13:67. [PMID: 32499677 PMCID: PMC7242790 DOI: 10.3389/fnmol.2020.00067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022] Open
Abstract
Dopamine, alongside other neuromodulators, defines brain and neuronal states, inter alia through regulation of global and local mRNA translation. Yet, the signaling pathways underlying the effects of dopamine on mRNA translation and psychiatric disorders are not clear. In order to examine the molecular pathways downstream of dopamine receptors, we used genetic, pharmacologic, biochemical, and imaging methods, and found that activation of dopamine receptor D1 but not D2 leads to rapid dephosphorylation of eEF2 at Thr56 but not eIF2α in cortical primary neuronal culture in a time-dependent manner. NMDA receptor, mTOR, and ERK pathways are upstream of the D1 receptor-dependent eEF2 dephosphorylation and essential for it. Furthermore, D1 receptor activation resulted in a major reduction in dendritic eEF2 phosphorylation levels. D1-dependent eEF2 dephosphorylation results in an increase of BDNF and synapsin2b expression which was followed by a small yet significant increase in general protein synthesis. These results reveal the role of dopamine D1 receptor in the regulation of eEF2 pathway translation in neurons and present eEF2 as a promising therapeutic target for addiction and depression as well as other psychiatric disorders.
Collapse
Affiliation(s)
- Orit David
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Iliana Barrera
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel.,School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Nathaniel Gould
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | | | - Kobi Rosenblum
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel.,Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| |
Collapse
|
29
|
Liu Z, Peng C, Zhuang Y, Chen Y, Behnisch T. Direct Medial Entorhinal Cortex Input to Hippocampal CA3 Is Crucial for eEF2K Inhibitor-Induced Neuronal Oscillations in the Mouse Hippocampus. Front Cell Neurosci 2020; 14:24. [PMID: 32210764 PMCID: PMC7069380 DOI: 10.3389/fncel.2020.00024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/27/2020] [Indexed: 11/13/2022] Open
Abstract
The hippocampal formation plays a vital role in memory formation and takes part in the control of the default neuronal network activity of the brain. It also represents an important structure to analyze drug-induced effects on subregion-specific synchronization of neuronal activity. However, the consequences of an altered functional state of synapses for subregion-specific synchronization of neuronal microcircuits remain to be fully understood. Therefore, we analyzed the direct interaction of neuronal microcircuits utilizing a genetically encoded calcium sensor (GCaMP6s) and local field potential (LFP) recording in acute hippocampal-entorhinal brain slices in response to a modulator of synaptic transmission. We observed that application of the eukaryotic elongation factor-2 kinase (eEF2K) inhibitor A484954, induced a large-scale synchronization of neuronal activity within different regions of the hippocampal formation. This effect was confirmed by the recording of extracellular LFPs. Further, in order to understand if the synchronized activity depended on interconnected hippocampal areas, we lesioned adjacent regions from each other. These experiments identified the origin of A484954-induced synchronized activity in the hippocampal CA3 subfield localized near the hilus of the dentate gyrus. Remarkably, the synchronization of neuronal activity in the hippocampus required an intact connection with the medial entorhinal cortex (MEC). In line with this observation, we detected an increase in neuronal activity in the MEC area after application of A484954. In summary, inhibition of eEF2K alters the intrinsic activity of interconnected neuronal microcircuits dominated by the MEC-CA3 afferents.
Collapse
Affiliation(s)
- Ziyang Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Cheng Peng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yinghan Zhuang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Oh JH, Nam TJ, Choi YH. Capsosiphon fulvescens Glycoproteins Enhance Probiotics-Induced Cognitive Improvement in Aged Rats. Nutrients 2020; 12:E837. [PMID: 32245093 PMCID: PMC7146536 DOI: 10.3390/nu12030837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 12/23/2022] Open
Abstract
Aging-induced cognitive dysfunction can be regulated by probiotics through bidirectional communication with the brain. This study aimed to investigate whether Capsosiphon fulvescens glycoproteins (Cf-hGP) enhanced probiotic-induced improvement of memory in aged rats and the underlying mechanism in the dorsal hippocampus. Cf-hGP were isolated using lectin resin. Cf-hGP (15 mg/kg/day) and/or Lactobacillus plantarum (L. plantarum) (109 CFU/rat/day) were orally administered once a day for 4 weeks. Co-treatment with Cf-hGP and L. plantarum synergistically improved spatial memory in aged rats, which was overturned by functional blocks of brain-derived neurotrophic factor (BDNF) signaling. Increases in BDNF expression and nuclear factor erythroid 2-related factor 2 (Nrf2) phosphorylation were accompanied by mono- and/or co-administration in the dorsal hippocampus, while c-Jun N-terminal kinase (JNK) phosphorylation and glucose-regulated protein 78 expression were decreased. These synergistic effects were downregulated by blocks of BDNF/Nrf2-mediated signaling. In particular, co-treatment, not mono-treatment, reduced phosphorylation of eukaryotic elongation factor 2 (eEF2) regulated by eEF2 kinase and protein phosphatase 2A. Additionally, co-treatment downregulated the interaction between eEF2 kinase and JNK. These data demonstrated that cognitive impairment in aged rats was synergistically diminished by co-treatment with Cf-hGP and L. plantarum through BDNF-mediated regulation of Nrf2 and eEF2 signaling pathways in the dorsal hippocampus.
Collapse
Affiliation(s)
- Jeong Hwan Oh
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea; (J.H.O.); (T.-J.N.)
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea; (J.H.O.); (T.-J.N.)
| | - Youn Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea; (J.H.O.); (T.-J.N.)
- Department of Marine Bio-Materials & Aquaculture, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
31
|
Beretta S, Gritti L, Verpelli C, Sala C. Eukaryotic Elongation Factor 2 Kinase a Pharmacological Target to Regulate Protein Translation Dysfunction in Neurological Diseases. Neuroscience 2020; 445:42-49. [PMID: 32088293 DOI: 10.1016/j.neuroscience.2020.02.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 02/02/2023]
Abstract
Two major processes tightly regulate protein synthesis, the initiation of mRNA translation and elongation phase that mediates the movement of ribosomes along the mRNA. The elongation phase is a high energy-consuming process, and is mainly regulated by the eukaryotic elongation factor 2 kinase (eEF2K) activity that phosphorylates and inhibits eEF2, the only known substrate of the kinase. eEF2K activity is closely regulated by several signaling pathways because the translation elongation phase strongly influences the cellular energy demand and can change the expression of specific proteins in different tissues. An increasing number of recent findings link eEF2k over activation to an array of human diseases, such as atherosclerosis, pulmonary arterial hypertension, progression of solid tumors, and some major neurological disorders. Several neurological studies suggest that eEF2K is a valuable target in treating epilepsy, depression and major neurodegenerative diseases. Despite eEF2k is an ubiquitous and conserved protein, it has been proved that its deletion does not affect development in animal models and in general cell viability. Therefore, it is possible to postulate that inhibiting its function may not cause serious side effects. In addition, eEF2K is a peculiar kinase molecularly different from most of other mammalian kinases and new compounds that inhibit eEF2K should not necessarily interfere with other important protein kinases. In this review we will critically summarize the evidence supporting the role of the altered eEF2K/eEF2 pathway in defined neurological diseases and its implications in curing these diseases in animal models, and possibly in humans, by targeting eEF2K activity.
Collapse
Affiliation(s)
| | | | | | - Carlo Sala
- CNR Neuroscience Institute, Milano, Italy.
| |
Collapse
|
32
|
Cataloguing and Selection of mRNAs Localized to Dendrites in Neurons and Regulated by RNA-Binding Proteins in RNA Granules. Biomolecules 2020; 10:biom10020167. [PMID: 31978946 PMCID: PMC7072219 DOI: 10.3390/biom10020167] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
Spatiotemporal translational regulation plays a key role in determining cell fate and function. Specifically, in neurons, local translation in dendrites is essential for synaptic plasticity and long-term memory formation. To achieve local translation, RNA-binding proteins in RNA granules regulate target mRNA stability, localization, and translation. To date, mRNAs localized to dendrites have been identified by comprehensive analyses. In addition, mRNAs associated with and regulated by RNA-binding proteins have been identified using various methods in many studies. However, the results obtained from these numerous studies have not been compiled together. In this review, we have catalogued mRNAs that are localized to dendrites and are associated with and regulated by the RNA-binding proteins fragile X mental retardation protein (FMRP), RNA granule protein 105 (RNG105, also known as Caprin1), Ras-GAP SH3 domain binding protein (G3BP), cytoplasmic polyadenylation element binding protein 1 (CPEB1), and staufen double-stranded RNA binding proteins 1 and 2 (Stau1 and Stau2) in RNA granules. This review provides comprehensive information on dendritic mRNAs, the neuronal functions of mRNA-encoded proteins, the association of dendritic mRNAs with RNA-binding proteins in RNA granules, and the effects of RNA-binding proteins on mRNA regulation. These findings provide insights into the mechanistic basis of protein-synthesis-dependent synaptic plasticity and memory formation and contribute to future efforts to understand the physiological implications of local regulation of dendritic mRNAs in neurons.
Collapse
|
33
|
Marcatili M, Sala C, Dakanalis A, Colmegna F, D'Agostino A, Gambini O, Dell'Osso B, Benatti B, Conti L, Clerici M. Human induced pluripotent stem cells technology in treatment resistant depression: novel strategies and opportunities to unravel ketamine's fast-acting antidepressant mechanisms. Ther Adv Psychopharmacol 2020; 10:2045125320968331. [PMID: 33224469 PMCID: PMC7649879 DOI: 10.1177/2045125320968331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Approximately 30% of Major Depressive Disorder (MDD) patients develop treatment-resistant depression (TRD). Among the different causes that make TRD so challenging in both clinical and research contexts, major roles are played by the inadequate understanding of MDD pathophysiology and the limitations of current pharmacological treatments. Nevertheless, the field of psychiatry is facing exciting times. Combined with recent advances in genome editing techniques, human induced pluripotent stem cell (hiPSC) technology is offering novel and unique opportunities in both disease modelling and drug discovery. This technology has allowed innovative disease-relevant patient-specific in vitro models to be set up for many psychiatric disorders. Such models hold great potential in enhancing our understanding of MDD pathophysiology and overcoming many of the well-known practical limitations inherent to animal and post-mortem models. Moreover, the field is approaching the advent of (es)ketamine, a glutamate N-methyl-d-aspartate (NMDA) receptor antagonist, claimed as one of the first and exemplary agents with rapid (in hours) antidepressant effects, even in TRD patients. Although ketamine seems poised to transform the treatment of depression, its exact mechanisms of action are still unclear but greatly demanded, as the resulting knowledge may provide a model to understand the mechanisms behind rapid-acting antidepressants, which may lead to the discovery of novel compounds for the treatment of depression. After reviewing insights into ketamine's mechanisms of action (derived from preclinical animal studies) and depicting the current state of the art of hiPSC technology below, we will consider the implementation of an hiPSC technology-based TRD model for the study of ketamine's fast acting antidepressant mechanisms of action.
Collapse
Affiliation(s)
- Matteo Marcatili
- Psychiatric Department, San Gerardo Hospital, ASST Monza, Monza, Italy
| | - Carlo Sala
- National Research Council Neuroscience Institute, Milan, Italy
| | - Antonios Dakanalis
- Department of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
| | - Fabrizia Colmegna
- Psychiatric Department, San Gerardo Hospital, ASST Monza, Monza, Italy
| | - Armando D'Agostino
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Orsola Gambini
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Bernardo Dell'Osso
- Psychiatry Unit, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Beatrice Benatti
- Psychiatry Unit, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Luciano Conti
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology (CIBIO), Università degli Studi di Trento, Trento, Italy
| | - Massimo Clerici
- Psychiatric Department, San Gerardo Hospital, ASST Monza, Monza, Italy
| |
Collapse
|
34
|
Wijasa TS, Sylvester M, Brocke-Ahmadinejad N, Schwartz S, Santarelli F, Gieselmann V, Klockgether T, Brosseron F, Heneka MT. Quantitative proteomics of synaptosome S-nitrosylation in Alzheimer's disease. J Neurochem 2019; 152:710-726. [PMID: 31520481 DOI: 10.1111/jnc.14870] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022]
Abstract
Increasing evidence suggests that both synaptic loss and neuroinflammation constitute early pathologic hallmarks of Alzheimer's disease. A downstream event during inflammatory activation of microglia and astrocytes is the induction of nitric oxide synthase type 2, resulting in an increased release of nitric oxide and the post-translational S-nitrosylation of protein cysteine residues. Both early events, inflammation and synaptic dysfunction, could be connected if this excess nitrosylation occurs on synaptic proteins. In the long term, such changes could provide new insight into patho-mechanisms as well as biomarker candidates from the early stages of disease progression. This study investigated S-nitrosylation in synaptosomal proteins isolated from APP/PS1 model mice in comparison to wild type and NOS2-/- mice, as well as human control, mild cognitive impairment and Alzheimer's disease brain tissues. Proteomics data were obtained using an established protocol utilizing an isobaric mass tag method, followed by nanocapillary high performance liquid chromatography tandem mass spectrometry. Statistical analysis identified the S-nitrosylation sites most likely derived from an increase in nitric oxide (NO) in dependence of presence of AD pathology, age and the key enzyme NOS2. The resulting list of candidate proteins is discussed considering function, previous findings in the context of neurodegeneration, and the potential for further validation studies.
Collapse
Affiliation(s)
| | - Marc Sylvester
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | | | - Stephanie Schwartz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | | | - Volkmar Gieselmann
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Thomas Klockgether
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurology, University of Bonn, Bonn, Germany
| | | | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
35
|
Dickson DW, Baker MC, Jackson JL, DeJesus-Hernandez M, Finch NA, Tian S, Heckman MG, Pottier C, Gendron TF, Murray ME, Ren Y, Reddy JS, Graff-Radford NR, Boeve BF, Petersen RC, Knopman DS, Josephs KA, Petrucelli L, Oskarsson B, Sheppard JW, Asmann YW, Rademakers R, van Blitterswijk M. Extensive transcriptomic study emphasizes importance of vesicular transport in C9orf72 expansion carriers. Acta Neuropathol Commun 2019; 7:150. [PMID: 31594549 PMCID: PMC6781370 DOI: 10.1186/s40478-019-0797-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022] Open
Abstract
The majority of the clinico-pathological variability observed in patients harboring a repeat expansion in the C9orf72-SMCR8 complex subunit (C9orf72) remains unexplained. This expansion, which represents the most common genetic cause of frontotemporal lobar degeneration (FTLD) and motor neuron disease (MND), results in a loss of C9orf72 expression and the generation of RNA foci and dipeptide repeat (DPR) proteins. The C9orf72 protein itself plays a role in vesicular transport, serving as a guanine nucleotide exchange factor that regulates GTPases. To further elucidate the mechanisms underlying C9orf72-related diseases and to identify potential disease modifiers, we performed an extensive RNA sequencing study. We included individuals for whom frontal cortex tissue was available: FTLD and FTLD/MND patients with (n = 34) or without (n = 44) an expanded C9orf72 repeat as well as control subjects (n = 24). In total, 6706 genes were differentially expressed between these groups (false discovery rate [FDR] < 0.05). The top gene was C9orf72 (FDR = 1.41E-14), which was roughly two-fold lower in C9orf72 expansion carriers than in (disease) controls. Co-expression analysis revealed groups of correlated genes (modules) that were enriched for processes such as protein folding, RNA splicing, synaptic signaling, metabolism, and Golgi vesicle transport. Within our cohort of C9orf72 expansion carriers, machine learning uncovered interesting candidates associated with clinico-pathological features, including age at onset (vascular endothelial growth factor A [VEGFA]), C9orf72 expansion size (cyclin dependent kinase like 1 [CDKL1]), DPR protein levels (eukaryotic elongation factor 2 kinase [EEF2K]), and survival after onset (small G protein signaling modulator 3 [SGSM3]). Given the fact that we detected a module involved in vesicular transport in addition to a GTPase activator (SGSM3) as a potential modifier, our findings seem to suggest that the presence of a C9orf72 repeat expansion might hamper vesicular transport and that genes affecting this process may modify the phenotype of C9orf72-linked diseases.
Collapse
Affiliation(s)
- Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Matthew C. Baker
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Jazmyne L. Jackson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | | | - NiCole A. Finch
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Shulan Tian
- Department of Health Sciences Research, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Michael G. Heckman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Cyril Pottier
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Tania F. Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Melissa E. Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Yingxue Ren
- Department of Health Sciences Research, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Joseph S. Reddy
- Department of Health Sciences Research, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | | | - Bradley F. Boeve
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - David S. Knopman
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Keith A. Josephs
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Björn Oskarsson
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - John W. Sheppard
- Gianforte School of Computing, Montana State University, 357 Barnard Hall, Bozeman, MT 59717 USA
| | - Yan W. Asmann
- Department of Health Sciences Research, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Marka van Blitterswijk
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
36
|
Electroacupuncture Improves Synaptic Function in SAMP8 Mice Probably via Inhibition of the AMPK/eEF2K/eEF2 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8260815. [PMID: 31641367 PMCID: PMC6766673 DOI: 10.1155/2019/8260815] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/30/2019] [Accepted: 08/21/2019] [Indexed: 01/07/2023]
Abstract
Synaptic loss and dysfunction is associated with cognitive impairment in Alzheimer's disease (AD). Recent evidence indicates that the AMP-activated protein kinase (AMPK)/eukaryotic elongation factor-2 kinase (eEF2K)/eukaryotic elongation factor-2 (eEF2) pathway was implicated in synaptic plasticity in AD. Therapeutic strategies for AD treatment are currently limited. Here, we investigate the effects of electroacupuncture (EA) on synaptic function and the AMPK/eEF2K/eEF2 signaling pathway in male senescence-accelerated mouse-prone 8 (SAMP8) mice. Male 7-month-old SAMP8 and SAMR1 mice (senescence-accelerated mouse resistant 1) were randomly divided into 3 groups: SAMR1 control group (Rc), SAMP8 control group (Pc), and SAMP8 electroacupuncture group (Pe). The Pe group was treated with EA for 30 days. Transmission electron microscopy (TEM) was used to observe the structure of synapse. The protein and mRNA expression of synaptophysin (SYN) and postsynaptic density 95 (PSD95) was examined by immunohistochemistry, western blot, and real-time RT-PCR. The activity of AMPK and eEF2K was studied by western blot. Our results showed that EA ameliorated synaptic loss, increased the expression of SYN and PSD95, and inhibited AMPK activation and eEF2K activity. Collectively, these findings suggested that the mechanisms of EA improving synaptic function in AD may be associated with the inhibition of the AMPK/eEF2K/eEF2 signaling pathway.
Collapse
|
37
|
Park H, Kaang BK. Balanced actions of protein synthesis and degradation in memory formation. ACTA ACUST UNITED AC 2019; 26:299-306. [PMID: 31416903 PMCID: PMC6699412 DOI: 10.1101/lm.048785.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/02/2019] [Indexed: 11/24/2022]
Abstract
Storage of long-term memory requires not only protein synthesis but also protein degradation. In this article, we overview recent publications related to this issue, stressing that the balanced actions of protein synthesis and degradation are critical for long-term memory formation. We particularly focused on the brain-derived neurotrophic factor signaling that leads to protein synthesis; proteasome- and autophagy-dependent protein degradation that removes molecular constraints; the role of Fragile X mental retardation protein in translational suppression; and epigenetic modifications that control gene expression at the genomic level. Numerous studies suggest that an imbalance between protein synthesis and degradation leads to intellectual impairment and cognitive disorders.
Collapse
Affiliation(s)
- Hyungju Park
- Department of Structure and Function of Neural Network, Korea Brain Research Institute (KBRI), Daegu 41062, South Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, South Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
38
|
Zimmermann HR, Yang W, Beckelman BC, Kasica NP, Zhou X, Galli LD, Ryazanov AG, Ma T. Genetic removal of eIF2α kinase PERK in mice enables hippocampal L-LTP independent of mTORC1 activity. J Neurochem 2019; 146:133-144. [PMID: 29337352 DOI: 10.1111/jnc.14306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 01/27/2023]
Abstract
Characterization of the molecular signaling pathways underlying protein synthesis-dependent forms of synaptic plasticity, such as late long-term potentiation (L-LTP), can provide insights not only into memory expression/maintenance under physiological conditions but also potential mechanisms associated with the pathogenesis of memory disorders. Here, we report in mice that L-LTP failure induced by the mammalian (mechanistic) target of rapamycin complex 1 (mTORC1) inhibitor rapamycin is reversed by brain-specific genetic deletion of PKR-like ER kinase, PERK (PERK KO), a kinase for eukaryotic initiation factor 2α (eIF2α). In contrast, genetic removal of general control non-derepressible-2, GCN2 (GCN2 KO), another eIF2α kinase, or treatment of hippocampal slices with the PERK inhibitor GSK2606414, does not rescue rapamycin-induced L-LTP failure, suggesting mechanisms independent of eIF2α phosphorylation. Moreover, we demonstrate that phosphorylation of eukaryotic elongation factor 2 (eEF2) is significantly decreased in PERK KO mice but unaltered in GCN2 KO mice or slices treated with the PERK inhibitor. Reduction in eEF2 phosphorylation results in increased general protein synthesis, and thus could contribute to the mTORC1-independent L-LTP in PERK KO mice. We further performed experiments on mutant mice with genetic removal of eEF2K (eEF2K KO), the only known kinase for eEF2, and found that L-LTP in eEF2K KO mice is insensitive to rapamycin. These data, for the first time, connect reduction in PERK activity with the regulation of translation elongation in enabling L-LTP independent of mTORC1. Thus, our findings indicate previously unrecognized levels of complexity in the regulation of protein synthesis-dependent synaptic plasticity. Read the Editorial Highlight for this article on page 119. Cover Image for this issue: doi: 10.1111/jnc.14185.
Collapse
Affiliation(s)
- Helena R Zimmermann
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Wenzhong Yang
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Brenna C Beckelman
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nicole P Kasica
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Xueyan Zhou
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Lucas Dufresne Galli
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alexey G Ryazanov
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.,Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.,Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
39
|
Martinetz S, Meinung CP, Jurek B, von Schack D, van den Burg EH, Slattery DA, Neumann ID. De Novo Protein Synthesis Mediated by the Eukaryotic Elongation Factor 2 Is Required for the Anxiolytic Effect of Oxytocin. Biol Psychiatry 2019; 85:802-811. [PMID: 30826070 DOI: 10.1016/j.biopsych.2019.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND The neuropeptide oxytocin (OXT) mediates its actions, including anxiolysis, via its G protein-coupled OXT receptor. Within the paraventricular nucleus of the hypothalamus (PVN), OXT-induced anxiolysis is mediated, at least in part, via activation of the mitogen-activated protein kinase pathway following calcium influx through transient receptor potential cation channel subfamily V member 2 channels. In the periphery, OXT activates eukaryotic elongation factor 2 (eEF2), an essential mediator of protein synthesis. METHODS In order to study whether OXT activates eEF2 also in neurons to exert its anxiolytic properties in the PVN, we performed in vivo and cell culture experiments. RESULTS We demonstrate that OXT, in a protein kinase C-dependent manner, activates eEF2 both in a hypothalamic cell line and in vivo within the PVN. Next, we reveal that OXT stimulates de novo protein synthesis, while inhibition of protein synthesis within the PVN prevents the anxiolytic effect of OXT in male rats. Moreover, activation of eEF2 within the PVN conveyed an anxiolytic effect supporting a role of OXT-induced eEF2 activation and protein synthesis for its anxiolysis. Finally, we show that one of the proteins that is upregulated by OXT is the neuropeptide Y receptor 5. Infusion of a specific neuropeptide Y receptor 5 agonist into the PVN consequently led to decreased anxiety-related behavior, while pretreatment with a neuropeptide Y receptor 5 antagonist prevented the anxiolytic effect of OXT. CONCLUSIONS Taken together, these results show that OXT recruits several intracellular signaling cascades to induce protein synthesis, which mediates the anxiolytic effects of OXT within the PVN and suggests that eEF2 represents a novel target for anxiety-related disorders.
Collapse
Affiliation(s)
- Stefanie Martinetz
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Carl-Philipp Meinung
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Benjamin Jurek
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - David von Schack
- Biotherapeutics Clinical Research and Development, Precision Medicine, New York, New York
| | | | - David A Slattery
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
40
|
Delaidelli A, Jan A, Herms J, Sorensen PH. Translational control in brain pathologies: biological significance and therapeutic opportunities. Acta Neuropathol 2019; 137:535-555. [PMID: 30739199 DOI: 10.1007/s00401-019-01971-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA) translation is the terminal step in protein synthesis, providing a crucial regulatory checkpoint for this process. Translational control allows specific cell types to respond to rapid changes in the microenvironment or to serve specific functions. For example, neurons use mRNA transport to achieve local protein synthesis at significant distances from the nucleus, the site of RNA transcription. Altered expression or functions of the various components of the translational machinery have been linked to several pathologies in the central nervous system. In this review, we provide a brief overview of the basic principles of mRNA translation, and discuss alterations of this process relevant to CNS disease conditions, with a focus on brain tumors and chronic neurological conditions. Finally, synthesizing this knowledge, we discuss the opportunities to exploit the biology of altered mRNA translation for novel therapies in brain disorders, as well as how studying these alterations can shed new light on disease mechanisms.
Collapse
Affiliation(s)
- Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Asad Jan
- Department of Biomedicine, Aarhus Institute of Advanced Studies, Aarhus University, Høegh-Guldbergs Gade 6B, 8000, Aarhus C, Denmark
| | - Jochen Herms
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University Munich, Schillerstraße 44, 80336, Munich, Germany
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
41
|
Crowley NA, Magee SN, Feng M, Jefferson SJ, Morris CJ, Dao NC, Brockway DF, Luscher B. Ketamine normalizes binge drinking-induced defects in glutamatergic synaptic transmission and ethanol drinking behavior in female but not male mice. Neuropharmacology 2019; 149:35-44. [PMID: 30731135 DOI: 10.1016/j.neuropharm.2019.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/30/2019] [Accepted: 02/02/2019] [Indexed: 01/06/2023]
Abstract
Ketamine is a fast acting experimental antidepressant with significant therapeutic potential for emotional disorders such as major depressive disorder and alcohol use disorders. Of particular interest is binge alcohol use, which during intermittent withdrawal from drinking involves depressive-like symptoms reminiscent of major depressive disorder. Binge drinking has been successfully modeled in mice with the Drinking in the Dark (DID) paradigm, which involves daily access to 20% ethanol, for a limited duration and selectively during the dark phase of the circadian light cycle. Here we demonstrate that DID exposure reduces the cell surface expression of NMDA- and AMPA-type glutamate receptors in the prelimbic cortex (PLC) of female but not male mice, along with reduced activity of the mammalian target of rapamycin (mTOR) signaling pathway. Pretreatment with an acute subanesthetic dose of ketamine suppresses binge-like ethanol consumption in female but not male mice. Lastly, DID-exposure reduces spontaneous glutamatergic synaptic transmission in the PLC of both sexes, but synaptic transmission is rescued by ketamine selectively in female mice. Thus, ketamine may have therapeutic potential as an ethanol binge suppressing agent selectively in female subjects.
Collapse
Affiliation(s)
- Nicole A Crowley
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sarah N Magee
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
| | - Mengyang Feng
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sarah J Jefferson
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Christian J Morris
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Nigel C Dao
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA; Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA
| | - Dakota F Brockway
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
42
|
Beckelman BC, Yang W, Kasica NP, Zimmermann HR, Zhou X, Keene CD, Ryazanov AG, Ma T. Genetic reduction of eEF2 kinase alleviates pathophysiology in Alzheimer's disease model mice. J Clin Invest 2019; 129:820-833. [PMID: 30667373 PMCID: PMC6355242 DOI: 10.1172/jci122954] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022] Open
Abstract
Molecular signaling mechanisms underlying Alzheimer's disease (AD) remain unclear. Maintenance of memory and synaptic plasticity depend on de novo protein synthesis, dysregulation of which is implicated in AD. Recent studies showed AD-associated hyperphosphorylation of mRNA translation factor eukaryotic elongation factor 2 (eEF2), which results in inhibition of protein synthesis. We tested to determine whether suppression of eEF2 phosphorylation could improve protein synthesis capacity and AD-associated cognitive and synaptic impairments. Genetic reduction of the eEF2 kinase (eEF2K) in 2 AD mouse models suppressed AD-associated eEF2 hyperphosphorylation and improved memory deficits and hippocampal long-term potentiation (LTP) impairments without altering brain amyloid β (Aβ) pathology. Furthermore, eEF2K reduction alleviated AD-associated defects in dendritic spine morphology, postsynaptic density formation, de novo protein synthesis, and dendritic polyribosome assembly. Our results link eEF2K/eEF2 signaling dysregulation to AD pathophysiology and therefore offer a feasible therapeutic target.
Collapse
Affiliation(s)
- Brenna C. Beckelman
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Wenzhong Yang
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Nicole P. Kasica
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Helena R. Zimmermann
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Xueyan Zhou
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - C. Dirk Keene
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Alexey G. Ryazanov
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Tao Ma
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Physiology and Pharmacology, and
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
43
|
Catanese A, Garrido D, Walther P, Roselli F, Boeckers TM. Nutrient limitation affects presynaptic structures through dissociable Bassoon autophagic degradation and impaired vesicle release. J Cereb Blood Flow Metab 2018; 38:1924-1939. [PMID: 29972341 PMCID: PMC6259322 DOI: 10.1177/0271678x18786356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acute mismatch between metabolic requirements of neurons and nutrients/growth factors availability characterizes several neurological conditions such as traumatic brain injury, stroke and hypoglycemia. Although the effects of this mismatch have been investigated at cell biological level, the effects on synaptic structure and function are less clear. Since synaptic activity is the most energy-demanding neuronal function and it is directly linked to neuronal networks functionality, we have explored whether nutrient limitation (NL) affects the ultrastructure, function and composition of pre and postsynaptic terminals. We show that upon NL, presynaptic terminals show disorganized vesicle pools and reduced levels of the active zone protein Bassoon (but not of Piccolo). Moreover, NL triggers an impaired vesicle release, which is reversed by re-administration of glucose but not by the blockade of autophagic or proteasomal protein degradation. This reveals a dissociable correlation between presynaptic architecture and vesicle release, since restoring vesicle fusion does not necessarily depend from the rescue of Bassoon levels. Thus, our data show that the presynaptic compartment is highly sensitive to NL and the rescue of presynaptic function requires re-establishment of the metabolic supply rather than preventing local protein degradation.
Collapse
Affiliation(s)
- Alberto Catanese
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,2 International Graduate School in Molecular Medicine Ulm (IGradU), Ulm University, Ulm, Germany
| | - Débora Garrido
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,2 International Graduate School in Molecular Medicine Ulm (IGradU), Ulm University, Ulm, Germany
| | - Paul Walther
- 3 Electron Microscopy Institute, Ulm University, Ulm, Germany
| | - Francesco Roselli
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,4 Department of Neurology, Ulm University, Ulm, Germany
| | - Tobias M Boeckers
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| |
Collapse
|
44
|
Stefanik MT, Milovanovic M, Werner CT, Spainhour JCG, Wolf ME. Withdrawal From Cocaine Self-administration Alters the Regulation of Protein Translation in the Nucleus Accumbens. Biol Psychiatry 2018; 84:223-232. [PMID: 29622268 PMCID: PMC6054574 DOI: 10.1016/j.biopsych.2018.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cue-induced cocaine craving incubates during abstinence from cocaine self-administration. Expression of incubation ultimately depends on elevation of homomeric GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors in the nucleus accumbens (NAc). This adaptation requires ongoing protein translation for its maintenance. Aberrant translation is implicated in central nervous system diseases, but nothing is known about glutamatergic regulation of translation in the drug-naïve NAc or after incubation. METHODS NAc tissue was obtained from drug-naïve rats and from rats after 1 or >40 days of abstinence from extended-access cocaine or saline self-administration. Newly translated proteins were labeled using 35S-Met/Cys or puromycin. We compared basal overall translation and its regulation by metabotropic glutamate receptor 1 (mGlu1), mGlu5, and N-methyl-D-aspartate receptors (NMDARs) in drug-naïve, saline control, and cocaine rats, and we compared GluA1 and GluA2 translation by immunoprecipitating puromycin-labeled proteins. RESULTS In all groups, overall translation was unaltered by mGlu1 blockade (LY367385) but increased by mGlu5 blockade (MTEP). NMDAR blockade (AVP) increased overall translation in drug-naïve and saline control rats but not in cocaine/late withdrawal rats. Cocaine/late withdrawal rats exhibited greater translation of GluA1 (but not GluA2), which was not further affected by NMDAR blockade. CONCLUSIONS Our results suggest that increased GluA1 translation contributes to the elevated homomeric GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor levels in the NAc that mediate incubation. Additional contributions to incubation-related plasticity may result from loss of the braking influence on translation normally exerted by NMDARs. Apart from elucidating incubation-related adaptations, we found a suppressive effect of mGlu5 on NAc translation regardless of drug exposure, which is opposite to results obtained in the hippocampus and points to heterogeneity of translational regulation between brain regions.
Collapse
Affiliation(s)
- Michael T Stefanik
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - Mike Milovanovic
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - Craig T Werner
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - John C G Spainhour
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, Georgia
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois.
| |
Collapse
|
45
|
Jan A, Jansonius B, Delaidelli A, Bhanshali F, An YA, Ferreira N, Smits LM, Negri GL, Schwamborn JC, Jensen PH, Mackenzie IR, Taubert S, Sorensen PH. Activity of translation regulator eukaryotic elongation factor-2 kinase is increased in Parkinson disease brain and its inhibition reduces alpha synuclein toxicity. Acta Neuropathol Commun 2018; 6:54. [PMID: 29961428 PMCID: PMC6027557 DOI: 10.1186/s40478-018-0554-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 06/10/2018] [Indexed: 01/05/2023] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder and the leading neurodegenerative cause of motor disability. Pathologic accumulation of aggregated alpha synuclein (AS) protein in brain, and imbalance in the nigrostriatal system due to the loss of dopaminergic neurons in the substantia nigra- pars compacta, are hallmark features in PD. AS aggregation and propagation are considered to trigger neurotoxic mechanisms in PD, including mitochondrial deficits and oxidative stress. The eukaryotic elongation factor-2 kinase (eEF2K) mediates critical regulation of dendritic mRNA translation and is a crucial molecule in diverse forms of synaptic plasticity. Here we show that eEF2K activity, assessed by immuonohistochemical detection of eEF2 phosphorylation on serine residue 56, is increased in postmortem PD midbrain and hippocampus. Induction of aggressive, AS-related motor phenotypes in a transgenic PD M83 mouse model also increased brain eEF2K expression and activity. In cultures of dopaminergic N2A cells, overexpression of wild-type human AS or the A53T mutant increased eEF2K activity. eEF2K inhibition prevented the cytotoxicity associated with AS overexpression in N2A cells by improving mitochondrial function and reduced oxidative stress. Furthermore, genetic deletion of the eEF2K ortholog efk-1 in C. elegans attenuated human A53T AS induced defects in behavioural assays reliant on dopaminergic neuron function. These data suggest a role for eEF2K activity in AS toxicity, and support eEF2K inhibition as a potential target in reducing AS-induced oxidative stress in PD.
Collapse
|
46
|
Liu DC, Seimetz J, Lee KY, Kalsotra A, Chung HJ, Lu H, Tsai NP. Mdm2 mediates FMRP- and Gp1 mGluR-dependent protein translation and neural network activity. Hum Mol Genet 2018; 26:3895-3908. [PMID: 29016848 DOI: 10.1093/hmg/ddx276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/11/2017] [Indexed: 01/08/2023] Open
Abstract
Activating Group 1 (Gp1) metabotropic glutamate receptors (mGluRs), including mGluR1 and mGluR5, elicits translation-dependent neural plasticity mechanisms that are crucial to animal behavior and circuit development. Dysregulated Gp1 mGluR signaling has been observed in numerous neurological and psychiatric disorders. However, the molecular pathways underlying Gp1 mGluR-dependent plasticity mechanisms are complex and have been elusive. In this study, we identified a novel mechanism through which Gp1 mGluR mediates protein translation and neural plasticity. Using a multi-electrode array (MEA) recording system, we showed that activating Gp1 mGluR elevates neural network activity, as demonstrated by increased spontaneous spike frequency and burst activity. Importantly, we validated that elevating neural network activity requires protein translation and is dependent on fragile X mental retardation protein (FMRP), the protein that is deficient in the most common inherited form of mental retardation and autism, fragile X syndrome (FXS). In an effort to determine the mechanism by which FMRP mediates protein translation and neural network activity, we demonstrated that a ubiquitin E3 ligase, murine double minute-2 (Mdm2), is required for Gp1 mGluR-induced translation and neural network activity. Our data showed that Mdm2 acts as a translation suppressor, and FMRP is required for its ubiquitination and down-regulation upon Gp1 mGluR activation. These data revealed a novel mechanism by which Gp1 mGluR and FMRP mediate protein translation and neural network activity, potentially through de-repressing Mdm2. Our results also introduce an alternative way for understanding altered protein translation and brain circuit excitability associated with Gp1 mGluR in neurological diseases such as FXS.
Collapse
Affiliation(s)
- Dai-Chi Liu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program
| | - Joseph Seimetz
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology
| | - Auinash Kalsotra
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,Carl R.Woese Institute of Genomic Biology, University of Illinois, Champaign, IL 61801, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hua Lu
- Department of Biochemistry and Molecular Biology.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
47
|
A heterogeneous tRNA granule structure exhibiting rapid, bi-directional neuritic transport. Eur J Cell Biol 2018; 97:168-179. [PMID: 29482850 DOI: 10.1016/j.ejcb.2018.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/09/2018] [Accepted: 02/12/2018] [Indexed: 11/23/2022] Open
Abstract
mRNA translation is regulated by diverse mechanisms that converge at the initiation and elongation steps to determine the rate, profile, and localization of proteins synthesized. A consistently relevant feature of these mechanisms is the spatial re-distribution of translation machinery, a process of particular importance in neural cells. This process has, however, been largely overlooked with respect to its potential role in regulating the local concentration of cytoplasmic tRNAs, even as a multitude of data suggest that spatial regulation of the tRNA pool may help explain the remarkably high rates of peptide elongation. Here, we report that Cy3/Cy5-labeled bulk tRNAs transfected into neural cells distribute into granule-like structures - "tRNA granules" - that exhibit dynamic mixing of tRNAs between granules and rapid, bi-directional vectorial movement within neurites. Imaging of endogenous tRNAgly and tRNAlys by fluorescent in situ hybridization revealed a similar granular distribution of tRNAs in somata and neurites; this distribution was highly overlapping with granules imaged by introduction of exogenous Cy5-tRNAthr and Cy3-tRNAval. A subset of tRNA granules located in the cell body, neurite branch points and growth cones displayed fluorescence resonance energy transfer (FRET) between Cy3 and Cy5-labeled tRNAs indicative of translation, and co-localization with elongation machinery. A population of smaller, rapidly trafficked granules in neurites lacked FRET and showed poor colocalization with translation initiation and elongation factors, suggesting that they are a translationally inactive tRNA transport particle. Our data suggest that tRNAs are packaged into granules that are rapidly transported to loci where translation is needed, where they may greatly increase the local concentration of tRNAs in support of efficient elongation. The potential implications of this newly described structure for channeling of elongation, local translation, and diseases associated with altered tRNA levels or function are discussed.
Collapse
|
48
|
Tréfier A, Pellissier LP, Musnier A, Reiter E, Guillou F, Crépieux P. G Protein-Coupled Receptors As Regulators of Localized Translation: The Forgotten Pathway? Front Endocrinol (Lausanne) 2018; 9:17. [PMID: 29456523 PMCID: PMC5801404 DOI: 10.3389/fendo.2018.00017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/15/2018] [Indexed: 12/31/2022] Open
Abstract
G protein-coupled receptors (GPCRs) exert their physiological function by transducing a complex signaling network that coordinates gene expression and dictates the phenotype of highly differentiated cells. Much is known about the gene networks they transcriptionally regulate upon ligand exposure in a process that takes hours before a new protein is synthesized. However, far less is known about GPCR impact on the translational machinery and subsequent mRNA translation, although this gene regulation level alters the cell phenotype in a strikingly different timescale. In fact, mRNA translation is an early response kinetically connected to signaling events, hence it leads to the synthesis of a new protein within minutes following receptor activation. By these means, mRNA translation is responsive to subtle variations of the extracellular environment. In addition, when restricted to cell subcellular compartments, local mRNA translation contributes to cell micro-specialization, as observed in synaptic plasticity or in cell migration. The mechanisms that control where in the cell an mRNA is translated are starting to be deciphered. But how an extracellular signal triggers such local translation still deserves extensive investigations. With the advent of high-throughput data acquisition, it now becomes possible to review the current knowledge on the translatome that some GPCRs regulate, and how this information can be used to explore GPCR-controlled local translation of mRNAs.
Collapse
Affiliation(s)
- Aurélie Tréfier
- Biologie et Bioinformatique des Systèmes de Signalisation, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- IFCE, Nouzilly, France
| | - Lucie P. Pellissier
- Déficit de Récompense, GPCR et sociabilité, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- IFCE, Nouzilly, France
| | - Astrid Musnier
- Biologie et Bioinformatique des Systèmes de Signalisation, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- IFCE, Nouzilly, France
| | - Eric Reiter
- Biologie et Bioinformatique des Systèmes de Signalisation, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- IFCE, Nouzilly, France
| | - Florian Guillou
- Plasticité Génomique et Expression Phénotypique, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- IFCE, Nouzilly, France
| | - Pascale Crépieux
- Biologie et Bioinformatique des Systèmes de Signalisation, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247, Nouzilly, France
- Université François Rabelais, Tours, France
- IFCE, Nouzilly, France
- *Correspondence: Pascale Crépieux,
| |
Collapse
|
49
|
Engaging homeostatic plasticity to treat depression. Mol Psychiatry 2018; 23:26-35. [PMID: 29133952 DOI: 10.1038/mp.2017.225] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/11/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous mood disorder, making it difficult to develop a generalized, pharmacological therapy that is effective for all who suffer from MDD. Through the fortuitous discovery of N-methyl-D-aspartate receptor (NMDAR) antagonists as effective antidepressants, we have gained key insights into how antidepressant effects can be produced at the circuit and molecular levels. NMDAR antagonists act as rapid-acting antidepressants such that relief from depressive symptoms occurs within hours of a single injection. The mode of action of NMDAR antagonists seemingly relies on their ability to activate protein-synthesis-dependent homeostatic mechanisms that restore top-down excitatory connections. Recent evidence suggests that NMDAR antagonists relieve depressive symptoms by forming new synapses resulting in increased excitatory drive. This event requires the mammalian target of rapamycin complex 1 (mTORC1), a signaling pathway that regulates synaptic protein synthesis. Herein, we review critical studies that shed light on the action of NMDAR antagonists as rapid-acting antidepressants and how they engage a neuron's or neural network's homeostatic mechanisms to self-correct. Recent studies notably demonstrate that a shift in γ-amino-butyric acid receptor B (GABABR) function, from inhibitory to excitatory, is required for mTORC1-dependent translation with NMDAR antagonists. Finally, we discuss how GABABR activation of mTORC1 helps resolve key discrepancies between rapid-acting antidepressants and local homeostatic mechanisms.
Collapse
|
50
|
Ahmad F, Singh K, Das D, Gowaikar R, Shaw E, Ramachandran A, Rupanagudi KV, Kommaddi RP, Bennett DA, Ravindranath V. Reactive Oxygen Species-Mediated Loss of Synaptic Akt1 Signaling Leads to Deficient Activity-Dependent Protein Translation Early in Alzheimer's Disease. Antioxid Redox Signal 2017; 27:1269-1280. [PMID: 28264587 PMCID: PMC5655421 DOI: 10.1089/ars.2016.6860] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
AIMS Synaptic deficits are known to underlie the cognitive dysfunction seen in Alzheimer's disease (AD). Generation of reactive oxygen species (ROS) by β-amyloid has also been implicated in AD pathogenesis. However, it is unclear whether ROS contributes to synaptic dysfunction seen in AD pathogenesis and, therefore, we examined whether altered redox signaling could contribute to synaptic deficits in AD. RESULTS Activity dependent but not basal translation was impaired in synaptoneurosomes from 1-month old presymptomatic APPSwe/PS1ΔE9 (APP/PS1) mice, and this deficit was sustained till middle age (MA, 9-10 months). ROS generation leads to oxidative modification of Akt1 in the synapse and consequent reduction in Akt1-mechanistic target of rapamycin (mTOR) signaling, leading to deficiency in activity-dependent protein translation. Moreover, we found a similar loss of activity-dependent protein translation in synaptoneurosomes from postmortem AD brains. INNOVATION Loss of activity-dependent protein translation occurs presymptomatically early in the pathogenesis of AD. This is caused by ROS-mediated loss of pAkt1, leading to reduced synaptic Akt1-mTOR signaling and is rescued by overexpression of Akt1. ROS-mediated damage is restricted to the synaptosomes, indicating selectivity. CONCLUSIONS We demonstrate that ROS-mediated oxidative modification of Akt1 contributes to synaptic dysfunction in AD, seen as loss of activity-dependent protein translation that is essential for synaptic plasticity and maintenance. Therapeutic strategies promoting Akt1-mTOR signaling at synapses may provide novel target(s) for disease-modifying therapy in AD. Antioxid. Redox Signal. 27, 1269-1280.
Collapse
Affiliation(s)
- Faraz Ahmad
- 1 Centre for Neuroscience, Indian Institute of Science , Bangalore, India
| | - Kunal Singh
- 1 Centre for Neuroscience, Indian Institute of Science , Bangalore, India
| | - Debajyoti Das
- 1 Centre for Neuroscience, Indian Institute of Science , Bangalore, India
| | - Ruturaj Gowaikar
- 1 Centre for Neuroscience, Indian Institute of Science , Bangalore, India
| | - Eisha Shaw
- 1 Centre for Neuroscience, Indian Institute of Science , Bangalore, India
| | | | | | | | - David A Bennett
- 2 Rush Alzheimer's Disease Center, Rush University Medical Center , Chicago, Illinois
| | - Vijayalakshmi Ravindranath
- 1 Centre for Neuroscience, Indian Institute of Science , Bangalore, India .,3 Centre for Brain Research , Bangalore, India
| |
Collapse
|