1
|
Fu SJ, Cheng KM, Hsiao CT, Fang YC, Jeng CJ, Tang CY. Pin1 promotes human Ca V2.1 channel polyubiquitination by RNF138: pathophysiological implication for episodic ataxia type 2. Cell Commun Signal 2024; 22:571. [PMID: 39609819 PMCID: PMC11603662 DOI: 10.1186/s12964-024-01960-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024] Open
Abstract
Loss-of-function mutations in the human gene encoding the neuron-specific Ca2+ channel CaV2.1 are linked to the neurological disease episodic ataxia type 2 (EA2), as well as neurodevelopmental disorders such as developmental delay and developmental epileptic encephalopathy. Disease-associated CaV2.1 mutants may exhibit defective proteostasis and promote endoplasmic reticulum (ER)-associated degradation of their wild-type (WT) counterpart in a dominant-negative manner. The E3 ubiquitin ligase RNF138 was previously shown to mediate EA2-related aberrant degradation of CaV2.1 at the ER. Herein we aimed to elucidate the ER proteostasis mechanism of CaV2.1. The peptidyl-prolyl cis/trans isomerase, NIMA-interacting 1 (Pin1) was identified as a novel neuronal CaV2.1 binding partner that promoted polyubiquitination and proteasomal degradation of CaV2.1. Suppression of endogenous Pin1 level with either shRNA knockdown or the Pin1 inhibitor all-trans retinoic acid (ATRA) enhanced endogenous CaV2.1 protein level in neurons, and attenuated ER-associated degradation of CaV2.1 WT and EA2-causing mutants. Detailed mutation analyses suggested that Pin1 interacted with specific phosphorylated serine/threonine-proline motifs in the intracellular II-III loop and the distal carboxy-terminal region of human CaV2.1. We further generated Pin1-insensitive CaV2.1 constructs and demonstrated that, during ER quality control, Pin1 served as an upstream regulator of CaV2.1 polyubiquitination and degradation by RNF138. Pin1 regulation was required for the dominant-negative effect of EA2 missense mutants, but not nonsense mutants, on CaV2.1 WT protein expression. Our data are consistent with the idea that CaV2.1 proteostasis at the ER, as well as dominant-negative suppression of disease-causing loss-of-function mutants on CaV2.1 WT, entail both Pin1/RNF138-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Ssu-Ju Fu
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Kai-Min Cheng
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Cheng-Tsung Hsiao
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Neurology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ya-Ching Fang
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
2
|
Guptarak J, Scaduto P, Tumurbaatar B, Zhang WR, Jupiter D, Taglialatela G, Fracassi A. Cognitive integrity in Non-Demented Individuals with Alzheimer's Neuropathology is associated with preservation and remodeling of dendritic spines. Alzheimers Dement 2024; 20:4677-4691. [PMID: 38829680 PMCID: PMC11247701 DOI: 10.1002/alz.13900] [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: 01/26/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION Individuals referred to as Non-Demented with Alzheimer's Neuropathology (NDAN) exhibit cognitive resilience despite presenting Alzheimer's disease (AD) histopathological signs. Investigating the mechanisms behind this resilience may unveil crucial insights into AD resistance. METHODS DiI labeling technique was used to analyze dendritic spine morphology in control (CTRL), AD, and NDAN post mortem frontal cortex, particularly focusing on spine types near and far from amyloid beta (Aβ) plaques. RESULTS NDAN subjects displayed a higher spine density in regions distant from Aβ plaques versus AD patients. In distal areas from the plaques, NDAN individuals exhibited more immature spines, while AD patients had a prevalence of mature spines. Additionally, our examination of levels of Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1), a protein associated with synaptic plasticity and AD, showed significantly lower expression in AD versus NDAN and CTRL. DISCUSSION These results suggest that NDAN individuals undergo synaptic remodeling, potentially facilitated by Pin1, serving as a compensatory mechanism to preserve cognitive function despite AD pathology. HIGHLIGHTS Spine density is reduced near Aβ plaques compared to the distal area in CTRL, AD, and NDAN dendrites. NDAN shows higher spine density than AD in areas far from Aβ plaques. Far from Aβ plaques, NDAN has a higher density of immature spines, AD a higher density of mature spines. AD individuals show significantly lower levels of Pin1 compared to NDAN and CTRL.
Collapse
Affiliation(s)
- Jutatip Guptarak
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Pietro Scaduto
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Batbayar Tumurbaatar
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Wen Ru Zhang
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Daniel Jupiter
- Department of Biostatistics and Data Science, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Giulio Taglialatela
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Anna Fracassi
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| |
Collapse
|
3
|
Tsokas P, Hsieh C, Flores-Obando RE, Bernabo M, Tcherepanov A, Hernández AI, Thomas C, Bergold PJ, Cottrell JE, Kremerskothen J, Shouval HZ, Nader K, Fenton AA, Sacktor TC. KIBRA anchoring the action of PKMζ maintains the persistence of memory. SCIENCE ADVANCES 2024; 10:eadl0030. [PMID: 38924398 PMCID: PMC11204205 DOI: 10.1126/sciadv.adl0030] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
How can short-lived molecules selectively maintain the potentiation of activated synapses to sustain long-term memory? Here, we find kidney and brain expressed adaptor protein (KIBRA), a postsynaptic scaffolding protein genetically linked to human memory performance, complexes with protein kinase Mzeta (PKMζ), anchoring the kinase's potentiating action to maintain late-phase long-term potentiation (late-LTP) at activated synapses. Two structurally distinct antagonists of KIBRA-PKMζ dimerization disrupt established late-LTP and long-term spatial memory, yet neither measurably affects basal synaptic transmission. Neither antagonist affects PKMζ-independent LTP or memory that are maintained by compensating PKCs in ζ-knockout mice; thus, both agents require PKMζ for their effect. KIBRA-PKMζ complexes maintain 1-month-old memory despite PKMζ turnover. Therefore, it is not PKMζ alone, nor KIBRA alone, but the continual interaction between the two that maintains late-LTP and long-term memory.
Collapse
Affiliation(s)
- Panayiotis Tsokas
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Changchi Hsieh
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Rafael E. Flores-Obando
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Matteo Bernabo
- Department of Psychology, McGill University, Montreal, Quebec H3A 1G1, Canada
| | - Andrew Tcherepanov
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - A. Iván Hernández
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Christian Thomas
- Internal Medicine D (MedD), Department of Molecular Nephrology, University Hospital of Münster, 48149 Münster, Germany
| | - Peter J. Bergold
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - James E. Cottrell
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Joachim Kremerskothen
- Internal Medicine D (MedD), Department of Molecular Nephrology, University Hospital of Münster, 48149 Münster, Germany
| | - Harel Z. Shouval
- Department of Neurobiology and Anatomy, University of Texas Medical at Houston, Houston, TX 77030, USA
| | - Karim Nader
- Department of Psychology, McGill University, Montreal, Quebec H3A 1G1, Canada
| | - André A. Fenton
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
- Neuroscience Institute at NYU Langone Medical Center, New York, NY 10016, USA
| | - Todd C. Sacktor
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| |
Collapse
|
4
|
Gupta S, Khan J, Ghosh S. Molecular mechanism of cognitive impairment associated with Parkinson's disease: A stroke perspective. Life Sci 2024; 337:122358. [PMID: 38128756 DOI: 10.1016/j.lfs.2023.122358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/03/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Parkinson's disease (PD) is a common neurological illness that causes several motor and non-motor symptoms, most characteristically limb tremors and bradykinesia. PD is a slowly worsening disease that arises due to progressive neurodegeneration of specific areas of the brain, especially the substantia nigra of the midbrain. Even though PD has continuously been linked to a higher mortality risk in numerous epidemiologic studies, there have been significant discoveries regarding the connection between PD and stroke. The incidence of strokes such as cerebral infarction and hemorrhage is substantially associated with the development of PD. Moreover, cognitive impairments, primarily dementia, have been associated with stroke and PD. However, the underlying molecular mechanism of this phenomenon is still obscure. This concise review focuses on the relationship between stroke and PD, emphasizing the molecular mechanism of cognition deficit and memory loss evident in PD and stroke. Furthermore, we are also highlighting some potential drug molecules that can target both PD and stroke.
Collapse
Affiliation(s)
- Sanju Gupta
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India.
| |
Collapse
|
5
|
Malter JS. Pin1 and Alzheimer's disease. Transl Res 2023; 254:24-33. [PMID: 36162703 PMCID: PMC10111655 DOI: 10.1016/j.trsl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is an immense and growing public health crisis. Despite over 100 years of investigation, the etiology remains elusive and therapy ineffective. Despite current gaps in knowledge, recent studies have identified dysfunction or loss-of-function of Pin1, a unique cis-trans peptidyl prolyl isomerase, as an important step in AD pathogenesis. Here I review the functionality of Pin1 and its role in neurodegeneration.
Collapse
Affiliation(s)
- James S Malter
- Department of Pathology, UT Southwestern Medical Center, 5333 Harry Hines Blvd, Dallas, TX 75390.
| |
Collapse
|
6
|
Liu J. Involvement of PKMζ in Stress Response and Depression. Front Cell Neurosci 2022; 16:907767. [PMID: 35669107 PMCID: PMC9163780 DOI: 10.3389/fncel.2022.907767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
The stress system in the brain plays a pivotal role in keeping humans and animals from harmful stimuli. However, excessive stress will cause maladaptive changes to the stress system and lead to depression. Despite the high prevalence of depression, the treatment remains limited. PKMζ, an atypical PKC isoform, has been demonstrated to play a crucial role in maintaining long-term potentiation and memory. Recent evidence shows that PKMζ is also involved in stress response and depressive-like behavior. In particular, it was demonstrated that stress that resulted in depressive-like behavior could decrease the expression of PKMζ in the prefrontal cortex, which could be reversed by antidepressants. Importantly, modulation of PKMζ expression could regulate depressive-like behaviors and the actions of antidepressants. These data suggested that PKMζ could be a molecular target for developing novel antidepressants. Here, I review the advance on the role of PKMζ in mediating stress response and its involvement in the development of depression.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
7
|
Huertas MA, Newton AJH, McDougal RA, Sacktor TC, Shouval HZ. Conditions for Synaptic Specificity during the Maintenance Phase of Synaptic Plasticity. eNeuro 2022; 9:ENEURO.0064-22.2022. [PMID: 35443991 PMCID: PMC9087736 DOI: 10.1523/eneuro.0064-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/31/2022] [Indexed: 01/21/2023] Open
Abstract
Activity-dependent modifications of synaptic efficacies are a cellular substrate of learning and memory. Experimental evidence shows that these modifications are synapse specific and that the long-lasting effects are associated with the sustained increase in concentration of specific proteins like PKMζ However, such proteins are likely to diffuse away from their initial synaptic location and spread out to neighboring synapses, potentially compromising synapse specificity. In this article, we address the issue of synapse specificity during memory maintenance. Assuming that the long-term maintenance of synaptic plasticity is accomplished by a molecular switch, we carry out analytical calculations and perform simulations using the reaction-diffusion package in NEURON to determine the limits of synapse specificity during maintenance. Moreover, we explore the effects of the diffusion and degradation rates of proteins and of the geometrical characteristics of dendritic spines on synapse specificity. We conclude that the necessary conditions for synaptic specificity during maintenance require that molecular switches reside in dendritic spines. The requirement for synaptic specificity when the molecular switch resides in spines still imposes strong limits on the diffusion and turnover of rates of maintenance molecules, as well as on the morphologic properties of synaptic spines. These constraints are quite general and apply to most existing models suggested for maintenance. The parameter values can be experimentally evaluated, and if they do not fit the appropriate predicted range, the validity of this class of maintenance models would be challenged.
Collapse
Affiliation(s)
- Marco A Huertas
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston, TX 77030
| | - Adam J H Newton
- Yale Center for Medical Informatics, New Haven, CT 06520
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203
| | - Robert A McDougal
- Yale Center for Medical Informatics, New Haven, CT 06520
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520
| | - Todd Charlton Sacktor
- Department of Physiology, Pharmacology, Anesthesiology, and Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203
| | - Harel Z Shouval
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston, TX 77030
- Department Electrical and Computer Engineering, Rice University, Houston, TX 77005
| |
Collapse
|
8
|
Hou X, Yang F, Li A, Zhao D, Ma N, Chen L, Lin S, Lin Y, Wang L, Yan X, Zheng M, Lee TH, Zhou XZ, Lu KP, Liu H. The Pin1-CaMKII-AMPA Receptor Axis Regulates Epileptic Susceptibility. Cereb Cortex 2021; 31:3082-3095. [PMID: 33569579 DOI: 10.1093/cercor/bhab004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 10/28/2020] [Accepted: 12/27/2020] [Indexed: 12/11/2022] Open
Abstract
Pin1 is a unique isomerase that regulates protein conformation and function after phosphorylation. Pin1 aberration contributes to some neurological diseases, notably Alzheimer's disease, but its role in epilepsy is not fully understood. We found that Pin1-deficient mice had significantly increased seizure susceptibility in multiple chemical inducing models and developed age-dependent spontaneous epilepsy. Electrophysiologically, Pin1 ablation enhanced excitatory synaptic transmission to prefrontal cortex (PFC) pyramidal neurons without affecting their intrinsic excitability. Biochemically, Pin1 ablation upregulated AMPA receptors and GluA1 phosphorylation by acting on phosphorylated CaMKII. Clinically, Pin1 was decreased significantly, whereas phosphorylated CaMKII and GluA1 were increased in the neocortex of patients with epilepsy. Moreover, Pin1 expression restoration in the PFC of Pin1-deficient mice using viral gene transfer significantly reduced phosphorylated CaMKII and GluA1 and effectively suppressed their seizure susceptibility. Thus, Pin1-CaMKII-AMPA receptors are a novel axis controlling epileptic susceptibility, highlighting attractive new therapeutic strategies.
Collapse
Affiliation(s)
- Xiaojun Hou
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China.,Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Fan Yang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Angcheng Li
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Debao Zhao
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Nengjun Ma
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Linying Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China.,The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Suijin Lin
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Yuanxiang Lin
- The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Xingxue Yan
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Min Zheng
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Tae Ho Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Hekun Liu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| |
Collapse
|
9
|
Patel H, Zamani R. The role of PKMζ in the maintenance of long-term memory: a review. Rev Neurosci 2021; 32:481-494. [PMID: 33550786 DOI: 10.1515/revneuro-2020-0105] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/27/2020] [Indexed: 11/15/2022]
Abstract
Long-term memories are thought to be stored in neurones and synapses that undergo physical changes, such as long-term potentiation (LTP), and these changes can be maintained for long periods of time. A candidate enzyme for the maintenance of LTP is protein kinase M zeta (PKMζ), a constitutively active protein kinase C isoform that is elevated during LTP and long-term memory maintenance. This paper reviews the evidence and controversies surrounding the role of PKMζ in the maintenance of long-term memory. PKMζ maintains synaptic potentiation by preventing AMPA receptor endocytosis and promoting stabilisation of dendritic spine growth. Inhibition of PKMζ, with zeta-inhibitory peptide (ZIP), can reverse LTP and impair established long-term memories. However, a deficit of memory retrieval cannot be ruled out. Furthermore, ZIP, and in high enough doses the control peptide scrambled ZIP, was recently shown to be neurotoxic, which may explain some of the effects of ZIP on memory impairment. PKMζ knockout mice show normal learning and memory. However, this is likely due to compensation by protein-kinase C iota/lambda (PKCι/λ), which is normally responsible for induction of LTP. It is not clear how, or if, this compensatory mechanism is activated under normal conditions. Future research should utilise inducible PKMζ knockdown in adult rodents to investigate whether PKMζ maintains memory in specific parts of the brain, or if it represents a global memory maintenance molecule. These insights may inform future therapeutic targets for disorders of memory loss.
Collapse
Affiliation(s)
- Hamish Patel
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Reza Zamani
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| |
Collapse
|
10
|
Hsieh C, Tsokas P, Grau-Perales A, Lesburguères E, Bukai J, Khanna K, Chorny J, Chung A, Jou C, Burghardt NS, Denny CA, Flores-Obando RE, Hartley BR, Rodríguez Valencia LM, Hernández AI, Bergold PJ, Cottrell JE, Alarcon JM, Fenton AA, Sacktor TC. Persistent increases of PKMζ in memory-activated neurons trace LTP maintenance during spatial long-term memory storage. Eur J Neurosci 2021; 54:6795-6814. [PMID: 33540466 DOI: 10.1111/ejn.15137] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 01/28/2021] [Indexed: 01/12/2023]
Abstract
PKMζ is an autonomously active PKC isoform crucial for the maintenance of synaptic long-term potentiation (LTP) and long-term memory. Unlike other kinases that are transiently stimulated by second messengers, PKMζ is persistently activated through sustained increases in protein expression of the kinase. Therefore, visualizing increases in PKMζ expression during long-term memory storage might reveal the sites of its persistent action and thus the location of memory-associated LTP maintenance in the brain. Using quantitative immunohistochemistry validated by the lack of staining in PKMζ-null mice, we examined the amount and distribution of PKMζ in subregions of the hippocampal formation of wild-type mice during LTP maintenance and spatial long-term memory storage. During LTP maintenance in hippocampal slices, PKMζ increases in the pyramidal cell body and stimulated dendritic layers of CA1 for at least 2 hr. During spatial memory storage, PKMζ increases in CA1 pyramidal cells for at least 1 month, paralleling the persistence of the memory. During the initial expression of the memory, we tagged principal cells with immediate-early gene Arc promoter-driven transcription of fluorescent proteins. The subset of memory-tagged CA1 cells selectively increases expression of PKMζ during memory storage, and the increase persists in dendritic compartments within stratum radiatum for 1 month, indicating long-term storage of information in the CA3-to-CA1 pathway. We conclude that persistent increases in PKMζ trace the molecular mechanism of LTP maintenance and thus the sites of information storage within brain circuitry during long-term memory.
Collapse
Affiliation(s)
- Changchi Hsieh
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Panayiotis Tsokas
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | | | | | - Joseph Bukai
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Kunal Khanna
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Joelle Chorny
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Ain Chung
- Center for Neural Science, New York University, New York, New York, USA
| | - Claudia Jou
- Department of Psychology, Hunter College, The City University of New York, New York, NY, USA.,Department of Psychology, The Graduate Center, The City University of New York, New York, NY, USA
| | - Nesha S Burghardt
- Department of Psychology, Hunter College, The City University of New York, New York, NY, USA.,Department of Psychology, The Graduate Center, The City University of New York, New York, NY, USA
| | - Christine A Denny
- Department of Psychiatry, Columbia University Irving Medical Center, Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc, New York State Psychiatric Institute Kolb Research Annex, New York, NY, USA
| | - Rafael E Flores-Obando
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Benjamin Rush Hartley
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | | | - A Iván Hernández
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Peter J Bergold
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - James E Cottrell
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Juan Marcos Alarcon
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - André Antonio Fenton
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA.,Center for Neural Science, New York University, New York, New York, USA
| | - Todd Charlton Sacktor
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
11
|
Smolen P, Baxter DA, Byrne JH. Comparing Theories for the Maintenance of Late LTP and Long-Term Memory: Computational Analysis of the Roles of Kinase Feedback Pathways and Synaptic Reactivation. Front Comput Neurosci 2020; 14:569349. [PMID: 33390922 PMCID: PMC7772319 DOI: 10.3389/fncom.2020.569349] [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] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/16/2020] [Indexed: 11/26/2022] Open
Abstract
A fundamental neuroscience question is how memories are maintained from days to a lifetime, given turnover of proteins that underlie expression of long-term synaptic potentiation (LTP) or “tag” synapses as eligible for LTP. A likely solution relies on synaptic positive feedback loops, prominently including persistent activation of Ca2+/calmodulin kinase II (CaMKII) and self-activated synthesis of protein kinase M ζ (PKMζ). Data also suggest positive feedback based on recurrent synaptic reactivation within neuron assemblies, or engrams, is necessary to maintain memories. The relative importance of these mechanisms is controversial. To explore the likelihood that each mechanism is necessary or sufficient to maintain memory, we simulated maintenance of LTP with a simplified model incorporating persistent kinase activation, synaptic tagging, and preferential reactivation of strong synapses, and analyzed implications of recent data. We simulated three model variants, each maintaining LTP with one feedback loop: autonomous, self-activated PKMζ synthesis (model variant I); self-activated CamKII (model variant II); and recurrent reactivation of strengthened synapses (model variant III). Variant I predicts that, for successful maintenance of LTP, either 1) PKMζ contributes to synaptic tagging, or 2) a low constitutive tag level persists during maintenance independent of PKMζ, or 3) maintenance of LTP is independent of tagging. Variant II maintains LTP and suggests persistent CaMKII activation could maintain PKMζ activity, a feedforward interaction not previously considered. However, we note data challenging the CaMKII feedback loop. In Variant III synaptic reactivation drives, and thus predicts, recurrent or persistent activation of CamKII and other necessary kinases, plausibly contributing to persistent elevation of PKMζ levels. Reactivation is thus predicted to sustain recurrent rounds of synaptic tagging and incorporation of plasticity-related proteins. We also suggest (model variant IV) that synaptic reactivation and autonomous kinase activation could synergistically maintain LTP. We propose experiments that could discriminate these maintenance mechanisms.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States.,Engineering and Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - John H Byrne
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
12
|
Fagiani F, Govoni S, Racchi M, Lanni C. The Peptidyl-prolyl Isomerase Pin1 in Neuronal Signaling: from Neurodevelopment to Neurodegeneration. Mol Neurobiol 2020; 58:1062-1073. [PMID: 33083964 PMCID: PMC7878263 DOI: 10.1007/s12035-020-02179-8] [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: 06/24/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022]
Abstract
The peptidyl-prolyl isomerase Pin1 is a unique enzyme catalyzing the isomerization of the peptide bond between phosphorylated serine-proline or threonine-proline motifs in proteins, thereby regulating a wide spectrum of protein functions, including folding, intracellular signaling, transcription, cell cycle progression, and apoptosis. Pin1 has been reported to act as a key molecular switch inducing cell-type-specific effects, critically depending on the different phosphorylation patterns of its targets within different biological contexts. While its implication in proliferating cells, and, in particular, in the field of cancer, has been widely characterized, less is known about Pin1 biological functions in terminally differentiated and post-mitotic neurons. Notably, Pin1 is widely expressed in the central and peripheral nervous system, where it regulates a variety of neuronal processes, including neuronal development, apoptosis, and synaptic activity. However, despite studies reporting the interaction of Pin1 with neuronal substrates or its involvement in specific signaling pathways, a more comprehensive understanding of its biological functions at neuronal level is still lacking. Besides its implication in physiological processes, a growing body of evidence suggests the crucial involvement of Pin1 in aging and age-related and neurodegenerative diseases, including Alzheimer's disease, Parkinson disease, frontotemporal dementias, Huntington disease, and amyotrophic lateral sclerosis, where it mediates profoundly different effects, ranging from neuroprotective to neurotoxic. Therefore, a more detailed understanding of Pin1 neuronal functions may provide relevant information on the consequences of Pin1 deregulation in age-related and neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fagiani
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Viale Taramelli 14, 27100, Pavia, Italy.,Scuola Universitaria Superiore IUSS Pavia, P.zza Vittoria, 15, 27100, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Viale Taramelli 14, 27100, Pavia, Italy
| | - Marco Racchi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Viale Taramelli 14, 27100, Pavia, Italy
| | - Cristina Lanni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Viale Taramelli 14, 27100, Pavia, Italy.
| |
Collapse
|
13
|
Sharma M, Sajikumar S. G9a/GLP Complex Acts as a Bidirectional Switch to Regulate Metabotropic Glutamate Receptor-Dependent Plasticity in Hippocampal CA1 Pyramidal Neurons. Cereb Cortex 2020; 29:2932-2946. [PMID: 29982412 DOI: 10.1093/cercor/bhy161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/14/2018] [Accepted: 06/17/2018] [Indexed: 02/01/2023] Open
Abstract
Metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD) is conventionally considered to be solely dependent on local protein synthesis. Given the impact of epigenetics on memory, the intriguing question is whether epigenetic regulation influences mGluR-LTD as well. G9a/GLP histone lysine methyltransferase complex is crucial for brain development and goal-directed learning as well as for drug-addiction. In this study, we analyzed whether the epigenetic regulation by G9a/GLP complex affects mGluR-LTD in CA1 hippocampal pyramidal neurons of 5-7 weeks old male Wistar rats. In hippocampal slices with intact CA1 dendritic regions, inhibition of G9a/GLP activity abolished mGluR-LTD. The inhibition of this complex upregulated the expression of plasticity proteins like PKMζ, which mediated the prevention of mGluR-LTD expression by regulating the NSF-GluA2-mediated trafficking of AMPA receptors towards the postsynaptic site. G9a/GLP inhibition during the induction of mGluR-LTD also downregulated the protein levels of phosphorylated-GluA2 and Arc. Interestingly, G9a/GLP inhibition could not impede the mGluR-LTD when the cell-body was severed. Our study highlights the role of G9a/GLP complex in intact neuronal network as a bidirectional switch; when turned on, it facilitates the expression of mGluR-LTD, and when turned off, it promotes the expression of long-term potentiation.
Collapse
Affiliation(s)
- Mahima Sharma
- Department of Physiology, National University of Singapore, 2 Medical Drive, MD9, Singapore, Singapore.,Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, 2 Medical Drive, MD9, Singapore, Singapore.,Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore, Singapore
| |
Collapse
|
14
|
Delgado JY. An Alternative Pin1 Binding and Isomerization Site in the N-Terminus Domain of PSD-95. Front Mol Neurosci 2020; 13:31. [PMID: 32256312 PMCID: PMC7094161 DOI: 10.3389/fnmol.2020.00031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/11/2020] [Indexed: 11/13/2022] Open
Abstract
Phosphorylation-dependent peptidyl-prolyl cis-trans isomerization plays key roles in cell cycle progression, the pathogenesis of cancer, and age-related neurodegeneration. Most of our knowledge about the role of phosphorylation-dependent peptidyl-prolyl cis-trans isomerization and the enzyme catalyzing this reaction, the peptidyl-prolyl isomerase (Pin1), is largely limited to proteins not present in neurons. Only a handful of examples have shown that phosphorylation-dependent peptidyl-prolyl cis-trans isomerization, Pin1 binding, or Pin1-mediated peptidyl-prolyl cis-trans isomerization regulate proteins present at excitatory synapses. In this work, I confirm previous findings showing that Pin1 binds postsynaptic density protein-95 (PSD-95) and identify an alternative binding site in the phosphorylated N-terminus of the PSD-95. Pin1 associates via its WW domain with phosphorylated threonine (T19) and serine (S25) in the N-terminus domain of PSD-95 and this association alters the local conformation of PSD-95. Most importantly, I show that proline-directed phosphorylation of the N-terminus domain of PSD-95 alters the local conformation of this region. Therefore, proline-directed phosphorylation of the N-terminus of PSD-95, Pin1 association, and peptidyl-prolyl cis-trans isomerization may all play a role in excitatory synaptic function and synapse development.
Collapse
Affiliation(s)
- Jary Y Delgado
- Department of Neurobiology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
15
|
Helfer P, Shultz TR. A computational model of systems memory consolidation and reconsolidation. Hippocampus 2019; 30:659-677. [PMID: 31872960 DOI: 10.1002/hipo.23187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/05/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022]
Abstract
In the mammalian brain, newly acquired memories depend on the hippocampus (HPC) for maintenance and recall, but over time, the neocortex takes over these functions, rendering memories HPC-independent. The process responsible for this transformation is called systems memory consolidation. Reactivation of a well-consolidated memory can trigger a temporary return to a HPC-dependent state, a phenomenon known as systems memory reconsolidation. The neural mechanisms underlying systems memory consolidation and reconsolidation are not well understood. Here, we propose a neural model based on well-documented mechanisms of synaptic plasticity and stability and describe a computational implementation that demonstrates the model's ability to account for a range of findings from the systems consolidation and reconsolidation literature. We derive several predictions from the computational model and suggest experiments that may test its validity.
Collapse
Affiliation(s)
- Peter Helfer
- Department of Psychology, McGill University, 2001 McGill College, Montreal, QC, Canada
| | - Thomas R Shultz
- Department of Psychology, McGill University, 2001 McGill College, Montreal, QC, Canada
| |
Collapse
|
16
|
aPKC in neuronal differentiation, maturation and function. Neuronal Signal 2019; 3:NS20190019. [PMID: 32269838 PMCID: PMC7104321 DOI: 10.1042/ns20190019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
The atypical Protein Kinase Cs (aPKCs)—PRKCI, PRKCZ and PKMζ—form a subfamily within the Protein Kinase C (PKC) family. These kinases are expressed in the nervous system, including during its development and in adulthood. One of the aPKCs, PKMζ, appears to be restricted to the nervous system. aPKCs are known to play a role in a variety of cellular responses such as proliferation, differentiation, polarity, migration, survival and key metabolic functions such as glucose uptake, that are critical for nervous system development and function. Therefore, these kinases have garnered a lot of interest in terms of their functional role in the nervous system. Here we review the expression and function of aPKCs in neural development and in neuronal maturation and function. Despite seemingly paradoxical findings with genetic deletion versus gene silencing approaches, we posit that aPKCs are likely candidates for regulating many important neurodevelopmental and neuronal functions, and may be associated with a number of human neuropsychiatric diseases.
Collapse
|
17
|
Experiments with Snails Add to Our Knowledge about the Role of aPKC Subfamily Kinases in Learning. Int J Mol Sci 2019; 20:ijms20092117. [PMID: 31035721 PMCID: PMC6539039 DOI: 10.3390/ijms20092117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/28/2022] Open
Abstract
Protein kinase Mζ is considered important for memory formation and maintenance in different species, including invertebrates. PKMζ participates in multiple molecular pathways in neurons, regulating translation initiation rate, AMPA receptors turnover, synaptic scaffolding assembly, and other processes. Here, for the first time, we established the sequence of mRNA encoding PKMζ homolog in land snail Helix lucorum. We annotated important features of this mRNA: domains, putative capping sites, translation starts, and splicing sites. We discovered that this mRNA has at least two isoforms, and one of them lacks sequence encoding C1 domain. C1 deletion may be unique for snail because it has not been previously found in other species. We performed behavioral experiments with snails, measured expression levels of identified isoforms, and confirmed that their expression correlates with one type of learning.
Collapse
|
18
|
Smolen P, Baxter DA, Byrne JH. How can memories last for days, years, or a lifetime? Proposed mechanisms for maintaining synaptic potentiation and memory. ACTA ACUST UNITED AC 2019; 26:133-150. [PMID: 30992383 PMCID: PMC6478248 DOI: 10.1101/lm.049395.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
With memory encoding reliant on persistent changes in the properties of synapses, a key question is how can memories be maintained from days to months or a lifetime given molecular turnover? It is likely that positive feedback loops are necessary to persistently maintain the strength of synapses that participate in encoding. Such feedback may occur within signal-transduction cascades and/or the regulation of translation, and it may occur within specific subcellular compartments or within neuronal networks. Not surprisingly, numerous positive feedback loops have been proposed. Some posited loops operate at the level of biochemical signal-transduction cascades, such as persistent activation of Ca2+/calmodulin kinase II (CaMKII) or protein kinase Mζ. Another level consists of feedback loops involving transcriptional, epigenetic and translational pathways, and autocrine actions of growth factors such as BDNF. Finally, at the neuronal network level, recurrent reactivation of cell assemblies encoding memories is likely to be essential for late maintenance of memory. These levels are not isolated, but linked by shared components of feedback loops. Here, we review characteristics of some commonly discussed feedback loops proposed to underlie the maintenance of memory and long-term synaptic plasticity, assess evidence for and against their necessity, and suggest experiments that could further delineate the dynamics of these feedback loops. We also discuss crosstalk between proposed loops, and ways in which such interaction can facilitate the rapidity and robustness of memory formation and storage.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
19
|
Preparation of Synaptoneurosomes for the Study of Glutamate Receptor Function. Methods Mol Biol 2019. [PMID: 30707435 DOI: 10.1007/978-1-4939-9077-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The use of synaptoneurosomes (SN) enables the detection of synaptic activity including the assessment of glutamate receptor function. SN are normally prepared by filtration and centrifugation methods. Here we review the preparation of SN by Percoll density gradient methodology for downstream applications that assesses glutamate receptor function such as measuring de novo protein synthesis. Major procedural steps include preparation of discontinuous Percoll-sucrose density gradients, collection of brain tissue, preparation of brain homogenates, isolation of synaptoneurosome bands from the discontinuous Percoll-sucrose gradients, and radiolabeling SN proteins. De novo protein synthesis can be reproducibly measured in SN prepared by this method.
Collapse
|
20
|
Baltaci SB, Mogulkoc R, Baltaci AK. Molecular Mechanisms of Early and Late LTP. Neurochem Res 2019; 44:281-296. [PMID: 30523578 DOI: 10.1007/s11064-018-2695-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/31/2018] [Accepted: 12/04/2018] [Indexed: 12/01/2022]
Abstract
LTP is the most intensively studied cellular model of the memory and generally divided at least two distinct phases as early and late. E-LTP requires activation of CaMKII that initiates biochemical events and trafficking of proteins, which eventually potentiate synaptic transmission, and is independent of de novo protein synthesis. In contrast, L-LTP requires gene expression and local protein synthesis regulated via TrkB receptor- and functional prions CPEB2-3-mediated translation. Maintenance of LTP for longer periods depends on constitutively active PKMζ. Throughout this review, current knowledge about early and late phases of LTP will be reviewed.
Collapse
Affiliation(s)
- Saltuk Bugra Baltaci
- Faculty of Medicine, Department of Physiology, Selcuk University, 42031, Konya, Turkey
| | - Rasim Mogulkoc
- Faculty of Medicine, Department of Physiology, Selcuk University, 42031, Konya, Turkey
| | | |
Collapse
|
21
|
Bear MF, Cooke SF, Giese KP, Kaang BK, Kennedy MB, Kim JI, Morris RGM, Park P. In memoriam: John Lisman - commentaries on CaMKII as a memory molecule. Mol Brain 2018; 11:76. [PMID: 30593282 PMCID: PMC6309094 DOI: 10.1186/s13041-018-0419-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/24/2018] [Indexed: 11/10/2022] Open
Abstract
Shortly before he died in October 2017, John Lisman submitted an invited review to Molecular Brain on 'Criteria for identifying the molecular basis of the engram (CaMKII, PKMζ)'. John had no opportunity to read the referees' comments, and as a mark of the regard in which he was held by the neuroscience community the Editors decided to publish his review as submitted. This obituary takes the form of a series of commentaries on Lisman's review. At the same time we are publishing as a separate article a longer response by Todd Sacktor and André Fenton entitled 'What does LTP tell us about the roles of CaMKII and PKMζ in memory?' which presents the case for a rival memory molecule, PKMζ.
Collapse
Affiliation(s)
- Mark F. Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Sam F. Cooke
- King’s College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, SE5 8AF UK
| | - Karl Peter Giese
- King’s College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, SE5 8AF UK
| | - Bong-Kiun Kaang
- Department of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Mary B. Kennedy
- The Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
| | - Ji-il Kim
- Department of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Richard G. M. Morris
- Laboratory for Cognitive Neuroscience, Centre for Discovery Brain Sciences, Edinburgh Neuroscience, Edinburgh, EH8 9JZ UK
| | - Pojeong Park
- Department of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| |
Collapse
|
22
|
Shimizu T, Kanai K, Sugawara Y, Uchida C, Uchida T. Prolyl Isomerase Pin1 Directly Regulates Calcium/Calmodulin-Dependent Protein Kinase II Activity in Mouse Brains. Front Pharmacol 2018; 9:1351. [PMID: 30532705 PMCID: PMC6265371 DOI: 10.3389/fphar.2018.01351] [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] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/05/2018] [Indexed: 11/28/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) is abundant in the brain and functions as a mediator of calcium signaling. We found that the relative activity of CaMKII was significantly lower in the WT mouse brains than in the Pin1-/- mouse brains. Pin1 binds to phosphorylated CaMKII and weakens its activity. For this reason, the phosphorylation level of tau in the presence of Pin1 is lower than that in the absence of Pin1, and microtubule polymerization is not downregulated by CaMKII when Pin1 is present. These results suggest a novel mechanism of action of Pin1 to prevent neurodegeneration.
Collapse
Affiliation(s)
- Taiki Shimizu
- Molecular Enzymology, Department of Molecular Cell Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kenta Kanai
- Molecular Enzymology, Department of Molecular Cell Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yui Sugawara
- Molecular Enzymology, Department of Molecular Cell Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Chiyoko Uchida
- Department of Human Development and Culture, Fukushima University, Fukushima, Japan
| | - Takafumi Uchida
- Molecular Enzymology, Department of Molecular Cell Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
23
|
Infantile Amnesia Is Related to Developmental Immaturity of the Maintenance Mechanisms for Long-Term Potentiation. Mol Neurobiol 2018; 56:907-919. [PMID: 29804230 DOI: 10.1007/s12035-018-1119-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/11/2018] [Indexed: 01/11/2023]
Abstract
Infantile amnesia (IA) refers to the inability of adults to recall episodic memories from infancy or early childhood. While several hypotheses have been proposed to explain the occurrence of IA, the neurobiological and molecular bases for this accelerated forgetting phenomenon remain elusive. Using hippocampus-dependent object-location memory and contextual fear conditioning tasks, we confirmed that infant mice trained at postnatal day 20 (P20) displayed deficits in long-term memory retention compared to adult (P60) mice. The percentage of CA1 pyramidal neurons expressing phosphorylated cAMP-responsive element-binding protein after fear conditioning was significantly lower in P20 than P60 mice. P20 mice exhibited attenuated basal excitatory synaptic transmission and early-phase long-term potentiation (E-LTP) at Schaffer collateral-CA1 synapses compared to P60 mice, but conversely, P20 mice have a greater susceptibility to induce time-dependent reversal of LTP by low-frequency afferent stimulation than P60 mice. The protein levels of GluN2B subunit of N-methyl-D-aspartate receptors (NMDARs), protein kinase Mζ (PKMζ), and protein phosphatase 2B (PP2B) in hippocampal CA1 region were significantly higher in P20 than P60 mice. We also found that the levels of calcium/calmodulin-dependent protein kinase II α autophosphorylation at Thr286, GluA1 phosphorylation at Ser831, and PKMζ protein biosynthesis occurred during the ensuing maintenance of E-LTP were significantly lower in P20 than P60 mice. Pharmacological blockade of GluN2B-containing NMDARs or PP2B effectively restored deficits of E-LTP and long-term memory retention observed in P20 mice. Altogether, these findings suggest that developmental immaturity of the maintenance mechanisms for E-LTP is linked to the occurrence of IA.
Collapse
|
24
|
Helfer P, Shultz TR. Coupled feedback loops maintain synaptic long-term potentiation: A computational model of PKMzeta synthesis and AMPA receptor trafficking. PLoS Comput Biol 2018; 14:e1006147. [PMID: 29813048 PMCID: PMC5993340 DOI: 10.1371/journal.pcbi.1006147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/08/2018] [Accepted: 04/16/2018] [Indexed: 01/13/2023] Open
Abstract
In long-term potentiation (LTP), one of the most studied types of neural plasticity, synaptic strength is persistently increased in response to stimulation. Although a number of different proteins have been implicated in the sub-cellular molecular processes underlying induction and maintenance of LTP, the precise mechanisms remain unknown. A particular challenge is to demonstrate that a proposed molecular mechanism can provide the level of stability needed to maintain memories for months or longer, in spite of the fact that many of the participating molecules have much shorter life spans. Here we present a computational model that combines simulations of several biochemical reactions that have been suggested in the LTP literature and show that the resulting system does exhibit the required stability. At the core of the model are two interlinked feedback loops of molecular reactions, one involving the atypical protein kinase PKMζ and its messenger RNA, the other involving PKMζ and GluA2-containing AMPA receptors. We demonstrate that robust bistability-stable equilibria both in the synapse's potentiated and unpotentiated states-can arise from a set of simple molecular reactions. The model is able to account for a wide range of empirical results, including induction and maintenance of late-phase LTP, cellular memory reconsolidation and the effects of different pharmaceutical interventions.
Collapse
Affiliation(s)
- Peter Helfer
- Department of Psychology and Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Thomas R. Shultz
- Department of Psychology and School of Computer Science, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Stallings NR, O'Neal MA, Hu J, Kavalali ET, Bezprozvanny I, Malter JS. Pin1 mediates Aβ 42-induced dendritic spine loss. Sci Signal 2018; 11:11/522/eaap8734. [PMID: 29559586 PMCID: PMC6136423 DOI: 10.1126/scisignal.aap8734] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Early-stage Alzheimer's disease is characterized by the loss of dendritic spines in the neocortex of the brain. This phenomenon precedes tau pathology, plaque formation, and neurodegeneration and likely contributes to synaptic loss, memory impairment, and behavioral changes in patients. Studies suggest that dendritic spine loss is induced by soluble, multimeric amyloid-β (Aβ42), which, through postsynaptic signaling, activates the protein phosphatase calcineurin. We investigated how calcineurin caused spine pathology and found that the cis-trans prolyl isomerase Pin1 was a critical downstream target of Aβ42-calcineurin signaling. In dendritic spines, Pin1 interacted with and was dephosphorylated by calcineurin, which rapidly suppressed its isomerase activity. Knockout of Pin1 or exposure to Aβ42 induced the loss of mature dendritic spines, which was prevented by exogenous Pin1. The calcineurin inhibitor FK506 blocked dendritic spine loss in Aβ42-treated wild-type cells but had no effect on Pin1-null neurons. These data implicate Pin1 in dendritic spine maintenance and synaptic loss in early Alzheimer's disease.
Collapse
Affiliation(s)
- Nancy R Stallings
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Melissa A O'Neal
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jie Hu
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ege T Kavalali
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
26
|
Kinases of eIF2a Switch Translation of mRNA Subset during Neuronal Plasticity. Int J Mol Sci 2017; 18:ijms18102213. [PMID: 29065505 PMCID: PMC5666893 DOI: 10.3390/ijms18102213] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022] Open
Abstract
Compared to other types of cells, neurons express the largest number of diverse mRNAs, including neuron-specific ones. This mRNA diversity is required for neuronal function, memory storage, maintenance and retrieval. Regulation of translation in neurons is very complicated and involves various proteins. Some proteins, implementing translational control in other cell types, are used by neurons for synaptic plasticity. In this review, we discuss the neuron-specific activity of four kinases: protein kinase R (PKR), PKR-like endoplasmic reticulum kinase (PERK), general control nonderepressible 2 kinase (GCN2), and heme-reguated eIF2α kinase (HRI), the substrate for which is α-subunit of eukaryotic initiation factor 2 (eIF2α). Phosphorylation of eIF2α is necessary for the cell during stress conditions, such as lack of amino acids, energy stress or viral infection. We propose that, during memory formation, neurons use some mechanisms similar to those involved in the cellular stress. The four eIF2α kinases regulate translation of certain mRNAs containing upstream open reading frames (uORFs). These mRNAs encode proteins involved in the processes of long-term potentiation (LTP) or long-term depression (LTD). The review examines some neuronal proteins for which translation regulation by eIF2 was suggested and checked experimentally. Of such proteins, we pay close attention to protein kinase Mζ, which is involved in memory storage and regulated at the translational level.
Collapse
|
27
|
Esnault S, Shen ZJ, Malter JS. Protein Translation and Signaling in Human Eosinophils. Front Med (Lausanne) 2017; 4:150. [PMID: 28971096 PMCID: PMC5609579 DOI: 10.3389/fmed.2017.00150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/01/2017] [Indexed: 01/01/2023] Open
Abstract
We have recently reported that, unlike IL-5 and GM-CSF, IL-3 induces increased translation of a subset of mRNAs. In addition, we have demonstrated that Pin1 controls the activity of mRNA binding proteins, leading to enhanced mRNA stability, GM-CSF protein production and prolonged eosinophil (EOS) survival. In this review, discussion will include an overview of cap-dependent protein translation and its regulation by intracellular signaling pathways. We will address the more general process of mRNA post-transcriptional regulation, especially regarding mRNA binding proteins, which are critical effectors of protein translation. Furthermore, we will focus on (1) the roles of IL-3-driven sustained signaling on enhanced protein translation in EOS, (2) the mechanisms regulating mRNA binding proteins activity in EOS, and (3) the potential targeting of IL-3 signaling and the signaling leading to mRNA binding activity changes to identify therapeutic targets to treat EOS-associated diseases.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine, Allergy, Pulmonary, and Critical Care Medicine Division, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Zhong-Jian Shen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
28
|
Abstract
Memory is an adaptation to particular temporal properties of past events, such as the frequency of occurrence of a stimulus or the coincidence of multiple stimuli. In neurons, this adaptation can be understood in terms of a hierarchical system of molecular and cellular time windows, which collectively retain information from the past. We propose that this system makes various timescales of past experience simultaneously available for future adjustment of behavior. More generally, we propose that the ability to detect and respond to temporally structured information underlies the nervous system's capacity to encode and store a memory at molecular, cellular, synaptic, and circuit levels.
Collapse
Affiliation(s)
| | - Thomas James Carew
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
29
|
ZIP It: Neural Silencing Is an Additional Effect of the PKM-Zeta Inhibitor Zeta-Inhibitory Peptide. J Neurosci 2017; 36:6193-8. [PMID: 27277798 DOI: 10.1523/jneurosci.4563-14.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/25/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Protein kinase M ζ (PKMζ), an atypical isoform of protein kinase C, has been suggested to be necessary and sufficient for the maintenance of long-term potentiation (LTP) and long-term memory (LTM). This evidence is heavily based on the use of ζ inhibitory peptide (ZIP), a supposed specific inhibitor of PKMζ that interferes with both LTP and LTM. Problematically, both LTP and LTM are unaffected in both constitutive and conditional PKMζ knock-out mice, yet both are still impaired by ZIP application, suggesting a nonspecific mechanism of action. Because translational interference can disrupt neural activity, we assessed network activity after a unilateral intrahippocampal infusion of ZIP in anesthetized rats. ZIP profoundly reduced spontaneous hippocampal local field potentials, comparable in magnitude to infusions of lidocaine, but with a slower onset and longer duration. Our results highlight a serious confound in interpreting the behavioral effects of ZIP. We suggest that future molecular approaches in neuroscience consider the intervening level of cellular and systems neurophysiology before claiming influences on behavior. SIGNIFICANCE STATEMENT Long-term memory in the brain is thought to arise from a sustained molecular process that can maintain changes in synaptic plasticity. A so-called candidate for the title of "the memory molecule" is protein kinase M ζ (PKMζ), mainly because its inhibition by ζ inhibitory peptide (ZIP) interferes with previously established synaptic plasticity and memory. We show that brain applications of ZIP that can impair memory actually profoundly suppress spontaneous brain activity directly or can cause abnormal seizure activity. We suggest that normal brain activity occurring after learning may be a more primary element of memory permanence.
Collapse
|
30
|
Pin1 Modulates the Synaptic Content of NMDA Receptors via Prolyl-Isomerization of PSD-95. J Neurosci 2017; 36:5437-47. [PMID: 27194325 DOI: 10.1523/jneurosci.3124-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 03/14/2016] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Phosphorylation of serine/threonine residues preceding a proline regulates the fate of its targets through postphosphorylation conformational changes catalyzed by the peptidyl-prolyl cis-/trans isomerase Pin1. By flipping the substrate between two different functional conformations, this enzyme exerts a fine-tuning of phosphorylation signals. Pin1 has been detected in dendritic spines and shafts where it regulates protein synthesis required to sustain the late phase of long-term potentiation (LTP). Here, we demonstrate that Pin1 residing in postsynaptic structures can interact with postsynaptic density protein-95 (PSD-95), a key scaffold protein that anchors NMDA receptors (NMDARs) in PSD via GluN2-type receptor subunits. Pin1 recruitment by PSD-95 occurs at specific serine-threonine/proline consensus motifs localized in the linker region connecting PDZ2 to PDZ3 domains. Upon binding, Pin1 triggers structural changes in PSD-95, thus negatively affecting its ability to interact with NMDARs. In electrophysiological experiments, larger NMDA-mediated synaptic currents, evoked in CA1 principal cells by Schaffer collateral stimulation, were detected in hippocampal slices obtained from Pin1(-/-) mice compared with controls. Similar results were obtained in cultured hippocampal cells expressing a PSD-95 mutant unable to undergo prolyl-isomerization, thus indicating that the action of Pin1 on PSD-95 is critical for this effect. In addition, an enhancement in spine density and size was detected in CA1 principal cells of Pin1(-/-) or in Thy-1GFP mice treated with the pharmacological inhibitor of Pin1 catalytic activity PiB.Our data indicate that Pin1 controls synaptic content of NMDARs via PSD-95 prolyl-isomerization and the expression of dendritic spines, both required for LTP maintenance. SIGNIFICANCE STATEMENT PSD-95, a membrane-associated guanylate kinase, is the major scaffolding protein at excitatory postsynaptic densities and a potent regulator of synaptic strength and plasticity. The activity of PSD-95 is tightly controlled by several post-translational mechanisms including proline-directed phosphorylation. This signaling cascade regulates the fate of its targets through postphosphorylation conformational modifications catalyzed by the peptidyl-prolyl cis-/trans isomerase Pin1. Here, we uncover a new role of Pin1 in glutamatergic signaling. By interacting with PSD-95, Pin1 dampens PSD-95 ability to complex with NMDARs, thus negatively affecting NMDAR signaling and spine morphology. Our findings further emphasize the emerging role of Pin1 as a key modulator of synaptic transmission.
Collapse
|
31
|
Pathological Role of Peptidyl-Prolyl Isomerase Pin1 in the Disruption of Synaptic Plasticity in Alzheimer's Disease. Neural Plast 2017; 2017:3270725. [PMID: 28458925 PMCID: PMC5385220 DOI: 10.1155/2017/3270725] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/12/2016] [Indexed: 01/18/2023] Open
Abstract
Synaptic loss is the structural basis for memory impairment in Alzheimer's disease (AD). While the underlying pathological mechanism remains elusive, it is known that misfolded proteins accumulate as β-amyloid (Aβ) plaques and hyperphosphorylated Tau tangles decades before the onset of clinical disease. The loss of Pin1 facilitates the formation of these misfolded proteins in AD. Pin1 protein controls cell-cycle progression and determines the fate of proteins by the ubiquitin proteasome system. The activity of the ubiquitin proteasome system directly affects the functional and structural plasticity of the synapse. We localized Pin1 to dendritic rafts and postsynaptic density (PSD) and found the pathological loss of Pin1 within the synapses of AD brain cortical tissues. The loss of Pin1 activity may alter the ubiquitin-regulated modification of PSD proteins and decrease levels of Shank protein, resulting in aberrant synaptic structure. The loss of Pin1 activity, induced by oxidative stress, may also render neurons more susceptible to the toxicity of oligomers of Aβ and to excitation, thereby inhibiting NMDA receptor-mediated synaptic plasticity and exacerbating NMDA receptor-mediated synaptic degeneration. These results suggest that loss of Pin1 activity could lead to the loss of synaptic plasticity in the development of AD.
Collapse
|
32
|
Borodinova AA, Zuzina AB, Balaban PM. Role of atypical protein kinases in maintenance of long-term memory and synaptic plasticity. BIOCHEMISTRY (MOSCOW) 2017; 82:243-256. [DOI: 10.1134/s0006297917030026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Hsieh C, Tsokas P, Serrano P, Hernández AI, Tian D, Cottrell JE, Shouval HZ, Fenton AA, Sacktor TC. Persistent increased PKMζ in long-term and remote spatial memory. Neurobiol Learn Mem 2017; 138:135-144. [PMID: 27417578 PMCID: PMC5501180 DOI: 10.1016/j.nlm.2016.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 07/04/2016] [Accepted: 07/09/2016] [Indexed: 02/02/2023]
Abstract
PKMζ is an autonomously active PKC isoform that is thought to maintain both LTP and long-term memory. Whereas persistent increases in PKMζ protein sustain the kinase's action in LTP, the molecular mechanism for the persistent action of PKMζ during long-term memory has not been characterized. PKMζ inhibitors disrupt spatial memory when introduced into the dorsal hippocampus from 1day to 1month after training. Therefore, if the mechanisms of PKMζ's persistent action in LTP maintenance and long-term memory were similar, persistent increases in PKMζ would last for the duration of the memory, far longer than most other learning-induced gene products. Here we find that spatial conditioning by aversive active place avoidance or appetitive radial arm maze induces PKMζ increases in dorsal hippocampus that persist from 1day to 1month, coinciding with the strength and duration of memory retention. Suppressing the increase by intrahippocampal injections of PKMζ-antisense oligodeoxynucleotides prevents the formation of long-term memory. Thus, similar to LTP maintenance, the persistent increase in the amount of autonomously active PKMζ sustains the kinase's action during long-term and remote spatial memory maintenance.
Collapse
Affiliation(s)
- Changchi Hsieh
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Panayiotis Tsokas
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Peter Serrano
- Department of Psychology, Hunter College, City University of New York, NY 10021, USA
| | - A Iván Hernández
- Department of Pathology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Dezhi Tian
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - James E Cottrell
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Harel Z Shouval
- Department of Neurobiology and Anatomy, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - André Antonio Fenton
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| | - Todd Charlton Sacktor
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA.
| |
Collapse
|
34
|
Bal NV, Susorov D, Chesnokova E, Kasianov A, Mikhailova T, Alkalaeva E, Balaban PM, Kolosov P. Upstream Open Reading Frames Located in the Leader of Protein Kinase Mζ mRNA Regulate Its Translation. Front Mol Neurosci 2016; 9:103. [PMID: 27790092 PMCID: PMC5061749 DOI: 10.3389/fnmol.2016.00103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/30/2016] [Indexed: 12/17/2022] Open
Abstract
For protein synthesis that occurs locally in dendrites, the translational control mechanisms are much more important for neuronal functioning than the transcription levels. Here, we show that uORFs (upstream open reading frames) in the 5′ untranslated region (5′UTR) play a critical role in regulation of the translation of protein kinase Mζ (PKMζ). Elimination of these uORFs activates translation of the reporter protein in vitro and in primary cultures of rat hippocampal neurons. Using cell-free translation systems, we demonstrate that translational initiation complexes are formed only on uORFs. Further, we address the mechanism of translational repression of PKMζ translation, by uORFs. We observed an increase in translation of the reporter protein under the control of PKMζ leader in neuronal culture during non-specific activation by picrotoxin. We also show that such a mechanism is similar to the mechanism seen in cell stress, as application of sodium arsenite to neuron cultures induced translation of mRNA carrying PKMζ 5′UTR similarly to picrotoxin activation. Therefore, we suppose that phosphorylation of eIF2a, like in cell stress, is a main regulator of PKMζ translation. Altogether, our findings considerably extend our understanding of the role of uORF in regulation of PKMζ translation in activated neurons, important at early stages of LTP.
Collapse
Affiliation(s)
- Natalia V Bal
- Cellular Neurobiology of Learning Laboratory, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences Moscow, Russia
| | - Denis Susorov
- Laboratory of Mechanisms and Control of Translation, Engelhardt Institute of Molecular Biology, Russian Academy of SciencesMoscow, Russia; Faculty of Bioengineering and Bioinformatics, M. V. Lomonosov Moscow State UniversityMoscow, Russia
| | - Ekaterina Chesnokova
- Cellular Neurobiology of Learning Laboratory, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences Moscow, Russia
| | - Artem Kasianov
- Laboratory of System Biology and Computational Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences Moscow, Russia
| | - Tatiana Mikhailova
- Laboratory of Mechanisms and Control of Translation, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences Moscow, Russia
| | - Elena Alkalaeva
- Laboratory of Mechanisms and Control of Translation, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences Moscow, Russia
| | - Pavel M Balaban
- Cellular Neurobiology of Learning Laboratory, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences Moscow, Russia
| | - Peter Kolosov
- Cellular Neurobiology of Learning Laboratory, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences Moscow, Russia
| |
Collapse
|
35
|
Overexpression of Protein Kinase Mζ in the Hippocampus Enhances Long-Term Potentiation and Long-Term Contextual But Not Cued Fear Memory in Rats. J Neurosci 2016; 36:4313-24. [PMID: 27076427 DOI: 10.1523/jneurosci.3600-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/17/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The persistently active protein kinase Mζ (PKMζ) has been found to be involved in the formation and maintenance of long-term memory. Most of the studies investigating PKMζ, however, have used either putatively unselective inhibitors or conventional knock-out animal models in which compensatory mechanisms may occur. Here, we overexpressed an active form of PKMζ in rat hippocampus, a structure highly involved in memory formation, and embedded in several neural networks. We investigated PKMζ's influence on synaptic plasticity using electrophysiological recordings of basal transmission, paired pulse facilitation, and LTP and combined this with behavioral cognitive experiments addressing formation and retention of both contextual memory during aversive conditioning and spatial memory during spontaneous exploration. We demonstrate that hippocampal slices overexpressing PKMζ show enhanced basal transmission, suggesting a potential role of PKMζ in postsynaptic AMPAR trafficking. Moreover, the PKMζ-overexpressing slices augmented LTP and this effect was not abolished by protein-synthesis blockers, indicating that PKMζ induces enhanced LTP formation in a protein-synthesis-independent manner. In addition, we found selectively enhanced long-term memory for contextual but not cued fear memory, underlining the theory of the hippocampus' involvement in the contextual aspect of aversive reinforced tasks. Memory for spatial orientation during spontaneous exploration remained unaltered, suggesting that PKMζ may not affect the neural circuits underlying spontaneous tasks that are different from aversive tasks. In this study, using an overexpression strategy as opposed to an inhibitor-based approach, we demonstrate an important modulatory role of PKMζ in synaptic plasticity and selective memory processing. SIGNIFICANCE STATEMENT Most of the literature investigating protein kinase Mζ (PKMζ) used inhibitors with selectivity that has been called into question or conventional knock-out animal models in which compensatory mechanisms may occur. To avoid these issues, some studies have been done using viral overexpression of PKMζ in different brain structures to show cognitive enhancement. However, electrophysiological experiments were exclusively done in knock-out models or inhibitory studies to show depletion of LTP. There was no study showing the effect of PKMζ overexpression in the hippocampus on behavior and LTP experiments. To our knowledge, this is the first study to combine these aspects with the result of enhanced memory for contextual fear memory and to show enhanced LTP in hippocampal slices overexpressing PKMζ.
Collapse
|
36
|
Sosa LJ, Malter JS, Hu J, Bustos Plonka F, Oksdath M, Nieto Guil AF, Quiroga S, Pfenninger KH. Protein interacting with NIMA (never in mitosis A)-1 regulates axonal growth cone adhesion and spreading through myristoylated alanine-rich C kinase substrate isomerization. J Neurochem 2016; 137:744-55. [DOI: 10.1111/jnc.13612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/20/2016] [Accepted: 03/03/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Lucas J. Sosa
- Department of Pediatrics and Colorado Intellectual and Developmental Disabilities Research Center; University of Colorado School of Medicine; Aurora Colorado USA
| | - James S. Malter
- Department of Pathology; University of Texas Southwestern Medical Center; Dallas Texas USA
| | - Jie Hu
- Department of Pathology; University of Texas Southwestern Medical Center; Dallas Texas USA
| | - Florentyna Bustos Plonka
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Mariana Oksdath
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Alvaro F. Nieto Guil
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Karl H. Pfenninger
- Department of Pediatrics and Colorado Intellectual and Developmental Disabilities Research Center; University of Colorado School of Medicine; Aurora Colorado USA
| |
Collapse
|
37
|
Jalil SJ, Sacktor TC, Shouval HZ. Atypical PKCs in memory maintenance: the roles of feedback and redundancy. ACTA ACUST UNITED AC 2015; 22:344-53. [PMID: 26077687 PMCID: PMC4478332 DOI: 10.1101/lm.038844.115] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/05/2015] [Indexed: 11/24/2022]
Abstract
Memories that last a lifetime are thought to be stored, at least in part, as persistent enhancement of the strength of particular synapses. The synaptic mechanism of these persistent changes, late long-term potentiation (L-LTP), depends on the state and number of specific synaptic proteins. Synaptic proteins, however, have limited dwell times due to molecular turnover and diffusion, leading to a fundamental question: how can this transient molecular machinery store memories lasting a lifetime? Because the persistent changes in efficacy are synapse-specific, the underlying molecular mechanisms must to a degree reside locally in synapses. Extensive experimental evidence points to atypical protein kinase C (aPKC) isoforms as key components involved in memory maintenance. Furthermore, it is evident that establishing long-term memory requires new protein synthesis. However, a comprehensive model has not been developed describing how these components work to preserve synaptic efficacies over time. We propose a molecular model that can account for key empirical properties of L-LTP, including its protein synthesis dependence, dependence on aPKCs, and synapse-specificity. Simulations and empirical data suggest that either of the two aPKC subtypes in hippocampal neurons, PKMζ and PKCι/λ, can maintain L-LTP, making the system more robust. Given genetic compensation at the level of synthesis of these PKC subtypes as in knockout mice, this system is able to maintain L-LTP and memory when one of the pathways is eliminated.
Collapse
Affiliation(s)
- Sajiya J Jalil
- Department of Neurobiology and Anatomy, The University of Texas Medical School at Houston, Houston, Texas 77030, USA
| | - Todd Charlton Sacktor
- Department of Physiology, Pharmacology, Anesthesiology, and Neurology, SUNY Downstate Medical Center, Brooklyn, New York 11203, USA
| | - Harel Z Shouval
- Department of Neurobiology and Anatomy, The University of Texas Medical School at Houston, Houston, Texas 77030, USA
| |
Collapse
|
38
|
Del Rosario JS, Feldmann KG, Ahmed T, Amjad U, Ko B, An J, Mahmud T, Salama M, Mei S, Asemota D, Mano I. Death Associated Protein Kinase (DAPK) -mediated neurodegenerative mechanisms in nematode excitotoxicity. BMC Neurosci 2015; 16:25. [PMID: 25899010 PMCID: PMC4414438 DOI: 10.1186/s12868-015-0158-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 03/31/2015] [Indexed: 12/30/2022] Open
Abstract
Background Excitotoxicity (the toxic overstimulation of neurons by the excitatory transmitter Glutamate) is a central process in widespread neurodegenerative conditions such as brain ischemia and chronic neurological diseases. Many mechanisms have been suggested to mediate excitotoxicity, but their significance across diverse excitotoxic scenarios remains unclear. Death Associated Protein Kinase (DAPK), a critical molecular switch that controls a range of key signaling and cell death pathways, has been suggested to have an important role in excitotoxicity. However, the molecular mechanism by which DAPK exerts its effect is controversial. A few distinct mechanisms have been suggested by single (sometimes contradicting) studies, and a larger array of potential mechanisms is implicated by the extensive interactome of DAPK. Results Here we analyze a well-characterized model of excitotoxicity in the nematode C. elegans to show that DAPK is an important mediator of excitotoxic neurodegeneration across a large evolutionary distance. We further show that some proposed mechanisms of DAPK’s action (modulation of synaptic strength, involvement of the DANGER-related protein MAB-21, and autophagy) do not have a major role in nematode excitotoxicity. In contrast, Pin1/PINN-1 (a DAPK interaction-partner and a peptidyl-prolyl isomerase involved in chronic neurodegenerative conditions) suppresses neurodegeneration in our excitotoxicity model. Conclusions Our studies highlight the prominence of DAPK and Pin1/PINN-1 as conserved mediators of cell death processes in diverse scenarios of neurodegeneration.
Collapse
Affiliation(s)
- John S Del Rosario
- Department of Physiology, Pharmacology, and Neuroscience, Sophie Davis School of Biomedical Education (SBE), City College of New York (CCNY), The City University of New York (CUNY), New York, NY, USA. .,MS program in Biology, CCNY, CUNY, New York, NY, USA.
| | - Katherine Genevieve Feldmann
- Department of Physiology, Pharmacology, and Neuroscience, Sophie Davis School of Biomedical Education (SBE), City College of New York (CCNY), The City University of New York (CUNY), New York, NY, USA. .,PhD program in Neuroscience, the CUNY Graduate Center, New York, NY, USA.
| | - Towfiq Ahmed
- Undergraduate program in Biology, CCNY, CUNY, New York, NY, USA.
| | - Uzair Amjad
- Undergraduate program in Biochemistry, CCNY, CUNY, New York, NY, USA.
| | - BakKeung Ko
- MS program in Biology, CCNY, CUNY, New York, NY, USA. .,Undergraduate program in Biology, CCNY, CUNY, New York, NY, USA.
| | - JunHyung An
- Undergraduate program in Biology, CCNY, CUNY, New York, NY, USA.
| | - Tauhid Mahmud
- Undergraduate program in Biology, CCNY, CUNY, New York, NY, USA.
| | - Maha Salama
- Bs/MD program, Sophie Davis SBE, CCNY, CUNY, New York, NY, USA.
| | - Shirley Mei
- Bs/MD program, Sophie Davis SBE, CCNY, CUNY, New York, NY, USA.
| | - Daniel Asemota
- Bs/MD program, Sophie Davis SBE, CCNY, CUNY, New York, NY, USA.
| | - Itzhak Mano
- Department of Physiology, Pharmacology, and Neuroscience, Sophie Davis School of Biomedical Education (SBE), City College of New York (CCNY), The City University of New York (CUNY), New York, NY, USA. .,PhD program in Neuroscience, the CUNY Graduate Center, New York, NY, USA.
| |
Collapse
|
39
|
Up-regulation of PKMζ expression in the anterior cingulate cortex following experimental tooth movement in rats. Arch Oral Biol 2014; 59:749-55. [DOI: 10.1016/j.archoralbio.2014.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/27/2014] [Accepted: 04/08/2014] [Indexed: 01/11/2023]
|
40
|
Zacchi P, Antonelli R, Cherubini E. Gephyrin phosphorylation in the functional organization and plasticity of GABAergic synapses. Front Cell Neurosci 2014; 8:103. [PMID: 24782709 PMCID: PMC3988358 DOI: 10.3389/fncel.2014.00103] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/22/2014] [Indexed: 11/13/2022] Open
Abstract
Gephyrin is a multifunctional scaffold protein essential for accumulation of inhibitory glycine and GABAA receptors at post-synaptic sites. The molecular events involved in gephyrin-dependent GABAA receptor clustering are still unclear. Evidence has been recently provided that gephyrin phosphorylation plays a key role in these processes. Gephyrin post-translational modifications have been shown to influence the structural remodeling of GABAergic synapses and synaptic plasticity by acting on post-synaptic scaffolding properties as well as stability. In addition, gephyrin phosphorylation and the subsequent phosphorylation-dependent recruitment of the chaperone molecule Pin1 provide a mechanism for the regulation of GABAergic signaling. Extensively characterized as pivotal enzyme controlling cell proliferation and differentiation, the prolyl-isomerase activity of Pin1 has been shown to regulate protein synthesis necessary to sustain the late phase of long-term potentiation at excitatory synapses, which suggests its involvement at synaptic sites. In this review we summarize the current state of knowledge of the signaling pathways responsible for gephyrin post-translational modifications. We will also outline future lines of research that might contribute to a better understanding of molecular mechanisms by which gephyrin regulates synaptic plasticity at GABAergic synapses.
Collapse
Affiliation(s)
- Paola Zacchi
- Department of Neurosciences, Scuola Internazionale Superiore di Studi Avanzati Trieste, Italy
| | - Roberta Antonelli
- Department of Neurosciences, Scuola Internazionale Superiore di Studi Avanzati Trieste, Italy
| | - Enrico Cherubini
- Department of Neurosciences, Scuola Internazionale Superiore di Studi Avanzati Trieste, Italy ; European Brain Research Institute Roma, Italy
| |
Collapse
|
41
|
Hernández AI, Oxberry WC, Crary JF, Mirra SS, Sacktor TC. Cellular and subcellular localization of PKMζ. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130140. [PMID: 24298142 DOI: 10.1098/rstb.2013.0140] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In contrast to protein kinases that participate in long-term potentiation (LTP) induction and memory consolidation, the autonomously active atypical protein kinase C isoform, protein kinase Mzeta (PKMζ), functions in the core molecular mechanism of LTP maintenance and long-term memory storage. Here, using multiple complementary techniques for light and electron microscopic immunolocalization, we present the first detailed characterization of the cellular and subcellular distribution of PKMζ in rat hippocampus and neocortex. We find that PKMζ is widely expressed in forebrain with prominent immunostaining in hippocampal and neocortical grey matter, and weak label in white matter. In hippocampal and cortical pyramidal cells, PKMζ expression is predominantly somatodendritic, and electron microscopy highlights the kinase at postsynaptic densities and in clusters within spines. In addition, nuclear label and striking punctate immunopositive structures in a paranuclear and dendritic distribution are seen by confocal microscopy, occasionally at dendritic bifurcations. PKMζ immunoreactive granules are observed by electron microscopy in cell bodies and dendrites, including endoplasmic reticulum. The widespread distribution of PKMζ in nuclei, nucleoli and endoplasmic reticulum suggests potential roles of this kinase in cell-wide mechanisms involving gene expression, biogenesis of ribosomes and new protein synthesis. The localization of PKMζ within postsynaptic densities and spines suggests sites where the kinase stores information during LTP maintenance and long-term memory.
Collapse
Affiliation(s)
- A Iván Hernández
- Department of Pathology, State University of New York, Downstate Medical Center, , Brooklyn, NY, USA
| | | | | | | | | |
Collapse
|
42
|
Hardt O, Nader K, Wang YT. GluA2-dependent AMPA receptor endocytosis and the decay of early and late long-term potentiation: possible mechanisms for forgetting of short- and long-term memories. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130141. [PMID: 24298143 DOI: 10.1098/rstb.2013.0141] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The molecular processes involved in establishing long-term potentiation (LTP) have been characterized well, but the decay of early and late LTP (E-LTP and L-LTP) is poorly understood. We review recent advances in describing the mechanisms involved in maintaining LTP and homeostatic plasticity. We discuss how these phenomena could relate to processes that might underpin the loss of synaptic potentiation over time, and how they might contribute to the forgetting of short-term and long-term memories. We propose that homeostatic downscaling mediates the loss of E-LTP, and that metaplastic parameters determine the decay rate of L-LTP, while both processes require the activity-dependent removal of postsynaptic GluA2-containing AMPA receptors.
Collapse
Affiliation(s)
- Oliver Hardt
- Centre for Cognitive and Neural Systems, University of Edinburgh, , Edinburgh, UK
| | | | | |
Collapse
|
43
|
Kwapis JL, Helmstetter FJ. Does PKM(zeta) maintain memory? Brain Res Bull 2013; 105:36-45. [PMID: 24076105 DOI: 10.1016/j.brainresbull.2013.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/16/2013] [Accepted: 09/19/2013] [Indexed: 11/30/2022]
Abstract
Work on the long-term stability of memory has identified a potentially critical role for protein kinase Mzeta (PKMζ) in maintaining established memory. PKMζ, an autonomously active isoform of PKC, is hypothesized to sustain those changes that occurred during memory formation in order to preserve the memory engram over time. Initial studies investigating the role of PKMζ were largely successful in demonstrating a role for the kinase in memory maintenance; disrupting PKMζ activity with ζ-inhibitory peptide (ZIP) was successful in disrupting a variety of established associations in a number of key brain regions. More recent work, however, has questioned both the role of PKMζ in memory maintenance and the effectiveness of ZIP as a specific inhibitor of PKMζ activity. Here, we outline the research both for and against the idea that PKMζ is a memory maintenance mechanism and discuss how these two lines of research can be reconciled. We conclude by proposing a number of studies that would help to clarify the role of PKMζ in memory and define other mechanisms the brain may use to maintain memory.
Collapse
Affiliation(s)
- Janine L Kwapis
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave., Milwaukee, WI 53211, USA
| | - Fred J Helmstetter
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave., Milwaukee, WI 53211, USA.
| |
Collapse
|
44
|
Panja D, Bramham CR. BDNF mechanisms in late LTP formation: A synthesis and breakdown. Neuropharmacology 2013; 76 Pt C:664-76. [PMID: 23831365 DOI: 10.1016/j.neuropharm.2013.06.024] [Citation(s) in RCA: 242] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 06/21/2013] [Accepted: 06/23/2013] [Indexed: 12/12/2022]
Abstract
Unraveling the molecular mechanisms governing long-term synaptic plasticity is a key to understanding how the brain stores information in neural circuits and adapts to a changing environment. Brain-derived neurotrophic factor (BDNF) has emerged as a regulator of stable, late phase long-term potentiation (L-LTP) at excitatory glutamatergic synapses in the adult brain. However, the mechanisms by which BDNF triggers L-LTP are controversial. Here, we distill and discuss the latest advances along three main lines: 1) TrkB receptor-coupled translational control underlying dendritic protein synthesis and L-LTP, 2) Mechanisms for BDNF-induced rescue of L-LTP when protein synthesis is blocked, and 3) BDNF-TrkB regulation of actin cytoskeletal dynamics in dendritic spines. Finally, we explore the inter-relationships between BDNF-regulated mechanisms, how these mechanisms contribute to different forms of L-LTP in the hippocampus and dentate gyrus, and outline outstanding issues for future research. This article is part of the Special Issue entitled 'BDNF Regulation of Synaptic Structure, Function, and Plasticity'.
Collapse
Affiliation(s)
- Debabrata Panja
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; KG Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | | |
Collapse
|
45
|
Oh J, Malter JS. Pin1-FADD interactions regulate Fas-mediated apoptosis in activated eosinophils. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:4937-45. [PMID: 23606538 PMCID: PMC3652414 DOI: 10.4049/jimmunol.1202646] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Abnormally long-lived eosinophils (Eos) are the major inflammatory component of allergic responses in the lungs of active asthmatics. Eos recruited to the airways after allergen exposure produce and respond to IL-5 and GM-CSF, enhancing their survival. Prosurvival signaling activates Pin1, a peptidyl-prolyl cis-trans isomerase that binds to Bax and prevents its activation. How long-lived Eos, despite the continued presence of GM-CSF or IL-5, eventually undergo apoptosis to end allergic inflammation remains unclear. In this study, we show that Pin1 location, activity, and protein interactions are jointly influenced by Fas and the prosurvival cytokine IL-5. Fas signaling strongly induced the phosphorylation of FADD at Ser(194) and Pin1 at Ser(16), as well as their nuclear accumulation. Phospho-mimic Ser(194)Glu FADD mutants accelerated Eos apoptosis compared with wild-type or Ser(194)Ala mutants. Downstream of FADD phosphorylation, caspase 8, 9, and 3 cleavage, as well as Eos apoptosis induced by Fas, were reduced by constitutively active Pin1 and enhanced by Pin1 inhibition. Pin1 was activated by IL-5, whereas simultaneous IL-5 and anti-Fas treatment modestly reduced peptidyl isomerase activity but induced Pin1 to associate with FADD after its phosphorylation at Ser(194). Mechanistically, Pin1-mediated isomerization facilitated the subsequent dephosphorylation of Ser(194) FADD and maintenance of cytoplasmic location. In vivo-activated bronchoalveolar Eos obtained after allergen challenge showed elevated survival and Pin1 activity that could be reversed by anti-Fas. Therefore, our data suggest that Pin1 is a critical link between FADD-mediated cell death and IL-5-mediated prosurvival signaling.
Collapse
Affiliation(s)
- Jiyoung Oh
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9072, USA
| | | |
Collapse
|
46
|
Swanger SA, Bassell GJ. Dendritic protein synthesis in the normal and diseased brain. Neuroscience 2012; 232:106-27. [PMID: 23262237 DOI: 10.1016/j.neuroscience.2012.12.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/21/2012] [Accepted: 12/01/2012] [Indexed: 01/25/2023]
Abstract
Synaptic activity is a spatially limited process that requires a precise, yet dynamic, complement of proteins within the synaptic micro-domain. The maintenance and regulation of these synaptic proteins is regulated, in part, by local mRNA translation in dendrites. Protein synthesis within the postsynaptic compartment allows neurons tight spatial and temporal control of synaptic protein expression, which is critical for proper functioning of synapses and neural circuits. In this review, we discuss the identity of proteins synthesized within dendrites, the receptor-mediated mechanisms regulating their synthesis, and the possible roles for these locally synthesized proteins. We also explore how our current understanding of dendritic protein synthesis in the hippocampus can be applied to new brain regions and to understanding the pathological mechanisms underlying varied neurological diseases.
Collapse
Affiliation(s)
- S A Swanger
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - G J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
47
|
Sacktor TC. Memory maintenance by PKMζ--an evolutionary perspective. Mol Brain 2012; 5:31. [PMID: 22986281 PMCID: PMC3517905 DOI: 10.1186/1756-6606-5-31] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/29/2012] [Indexed: 11/20/2022] Open
Abstract
Long-term memory is believed to be maintained by persistent modifications of synaptic transmission within the neural circuits that mediate behavior. Thus, long-term potentiation (LTP) is widely studied as a potential physiological basis for the persistent enhancement of synaptic strength that might sustain memory. Whereas the molecular mechanisms that initially induce LTP have been extensively characterized, the mechanisms that persistently maintain the potentiation have not. Recently, however, a candidate molecular mechanism linking the maintenance of LTP and the storage of long-term memory has been identified. The persistent activity of the autonomously active, atypical protein kinase C (aPKC) isoform, PKMζ, is both necessary and sufficient for maintaining LTP. Furthermore, blocking PKMζ activity by pharmacological or dominant negative inhibitors disrupts previously stored long-term memories in a variety of neural circuits, including spatial and trace memories in the hippocampus, aversive memories in the basolateral amygdala, appetitive memories in the nucleus accumbens, habit memory in the dorsal lateral striatum, and elementary associations, extinction, and skilled sensorimotor memories in the neocortex. During LTP and memory formation, PKMζ is synthesized de novo as a constitutively active kinase. This molecular mechanism for memory storage is evolutionarily conserved. PKMζ formation through new protein synthesis likely originated in early vertebrates ~500 million years ago during the Cambrian period. Other mechanisms for forming persistently active PKM from aPKC are found in invertebrates, and inhibiting this atypical PKM disrupts long-term memory in the invertebrate model systems Drosophila melanogaster and Aplysia californica. Conversely, overexpressing PKMζ enhances memory in flies and rodents. PKMζ persistently enhances synaptic strength by maintaining increased numbers of AMPA receptors at postsynaptic sites, a mechanism that might have evolved from the general function of aPKC in trafficking membrane proteins to the apical compartment of polarized cells. This mechanism of memory may have had adaptive advantages because it is both stable and reversible, as demonstrated by the downregulation of experience-dependent, long-term increases in PKMζ after extinction and reconsolidation blockade that attenuate learned behavior. Thus, PKMζ, the “working end” of LTP, is a component of an evolutionarily conserved molecular mechanism for the persistent, yet flexible storage of long-term memory.
Collapse
Affiliation(s)
- Todd Charlton Sacktor
- The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 10705, USA.
| |
Collapse
|
48
|
Long-term memory search across the visual brain. Neural Plast 2012; 2012:392695. [PMID: 22900206 PMCID: PMC3409559 DOI: 10.1155/2012/392695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 05/23/2012] [Accepted: 05/28/2012] [Indexed: 11/17/2022] Open
Abstract
Signal transmission from the human retina to visual cortex and connectivity of visual brain areas are relatively well understood. How specific visual perceptions transform into corresponding long-term memories remains unknown. Here, I will review recent Blood Oxygenation Level-Dependent functional Magnetic Resonance Imaging (BOLD fMRI) in humans together with molecular biology studies (animal models) aiming to understand how the retinal image gets transformed into so-called visual (retinotropic) maps. The broken object paradigm has been chosen in order to illustrate the complexity of multisensory perception of simple objects subject to visual —rather than semantic— type of memory encoding. The author explores how amygdala projections to the visual cortex affect the memory formation and proposes the choice of experimental techniques needed to explain our massive visual memory capacity. Maintenance of the visual long-term memories is suggested to require recycling of GluR2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) and β2-adrenoreceptors at the postsynaptic membrane, which critically depends on the catalytic activity of the N-ethylmaleimide-sensitive factor (NSF) and protein kinase PKMζ.
Collapse
|
49
|
Smolen P, Baxter DA, Byrne JH. Molecular constraints on synaptic tagging and maintenance of long-term potentiation: a predictive model. PLoS Comput Biol 2012; 8:e1002620. [PMID: 22876169 PMCID: PMC3410876 DOI: 10.1371/journal.pcbi.1002620] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 06/12/2012] [Indexed: 01/17/2023] Open
Abstract
Protein synthesis-dependent, late long-term potentiation (LTP) and depression (LTD) at glutamatergic hippocampal synapses are well characterized examples of long-term synaptic plasticity. Persistent increased activity of protein kinase M ζ (PKMζ) is thought essential for maintaining LTP. Additional spatial and temporal features that govern LTP and LTD induction are embodied in the synaptic tagging and capture (STC) and cross capture hypotheses. Only synapses that have been “tagged” by a stimulus sufficient for LTP and learning can “capture” PKMζ. A model was developed to simulate the dynamics of key molecules required for LTP and LTD. The model concisely represents relationships between tagging, capture, LTD, and LTP maintenance. The model successfully simulated LTP maintained by persistent synaptic PKMζ, STC, LTD, and cross capture, and makes testable predictions concerning the dynamics of PKMζ. The maintenance of LTP, and consequently of at least some forms of long-term memory, is predicted to require continual positive feedback in which PKMζ enhances its own synthesis only at potentiated synapses. This feedback underlies bistability in the activity of PKMζ. Second, cross capture requires the induction of LTD to induce dendritic PKMζ synthesis, although this may require tagging of a nearby synapse for LTP. The model also simulates the effects of PKMζ inhibition, and makes additional predictions for the dynamics of CaM kinases. Experiments testing the above predictions would significantly advance the understanding of memory maintenance. A fundamental problem in neurobiology is to understand how memories are maintained for up to years. Long-term potentiation (LTP), an enduring increase in the strength of specific connections (synapses) between neurons, is thought to comprise, at least in part, the substrate of learning and memory. What processes transduce brief stimuli into persistent LTP? Persistent increased activity of an enzyme denoted protein kinase M ζ (PKMζ) is thought essential for maintaining LTP. Only synapses that have been “tagged” by a stimulus, such as stimuli needed for LTP and learning, can “capture” PKMζ. We developed a model simulating dynamics of key molecules required for LTP and its opposite, long-term depression (LTD). The model concisely represents relationships between tagging, capture, LTD, and LTP maintenance. It makes testable predictions concerning the dynamics of PKMζ. The maintenance of LTP and memory is predicted to require positive feedback in which PKMζ enhances its own synthesis at potentiated synapses. Without synaptic capture of PKMζ, no positive feedback would occur. LTD induction is also predicted to increase PKMζ synthesis. The model also makes predictions about regulation of PKMζ synthesis. Experiments testing the above predictions would advance the understanding of memory maintenance.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, The University of Texas Medical School at Houston, Houston, Texas, United States of America.
| | | | | |
Collapse
|
50
|
Rangasamy V, Mishra R, Sondarva G, Das S, Lee TH, Bakowska JC, Tzivion G, Malter JS, Rana B, Lu KP, Kanthasamy A, Rana A. Mixed-lineage kinase 3 phosphorylates prolyl-isomerase Pin1 to regulate its nuclear translocation and cellular function. Proc Natl Acad Sci U S A 2012; 109:8149-54. [PMID: 22566623 PMCID: PMC3361382 DOI: 10.1073/pnas.1200804109] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nuclear protein peptidyl-prolyl isomerase Pin1-mediated prolyl isomerization is an essential and novel regulatory mechanism for protein phosphorylation. Therefore, tight regulation of Pin1 localization and catalytic activity is crucial for its normal nuclear functions. Pin1 is commonly dysregulated during oncogenesis and likely contributes to these pathologies; however, the mechanism(s) by which Pin1 catalytic activity and nuclear localization are increased is unknown. Here we demonstrate that mixed-lineage kinase 3 (MLK3), a MAP3K family member, phosphorylates Pin1 on a Ser138 site to increase its catalytic activity and nuclear translocation. This phosphorylation event drives the cell cycle and promotes cyclin D1 stability and centrosome amplification. Notably, Pin1 pSer138 is significantly up-regulated in breast tumors and is localized in the nucleus. These findings collectively suggest that the MLK3-Pin1 signaling cascade plays a critical role in regulating the cell cycle, centrosome numbers, and oncogenesis.
Collapse
Affiliation(s)
| | | | | | - Subhasis Das
- Departments of Molecular Pharmacology and Therapeutics and
| | - Tae Ho Lee
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | | | - Guri Tzivion
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216
| | - James S. Malter
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705
| | - Basabi Rana
- Medicine, Loyola University Chicago, Maywood, IL 60153
- Hines Veterans Affairs Medical Center, Hines, IL 60141; and
| | - Kun Ping Lu
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011
| | - Ajay Rana
- Departments of Molecular Pharmacology and Therapeutics and
- Hines Veterans Affairs Medical Center, Hines, IL 60141; and
| |
Collapse
|