Skip to main content

EDITORIAL article

Front. Synaptic Neurosci., 10 September 2024
This article is part of the Research Topic Role of Protein Palmitoylation in Synaptic Plasticity and Neuronal Differentiation, Volume II View all 5 articles

Editorial: Role of protein palmitoylation in synaptic plasticity and neuronal differentiation, volume II

  • Department of Neurobiology, University of Chicago, Chicago, IL, United States

Post-translational modifications (PTMs), such as S-palmitoylation described here, add to the already large diversity of protein amino acid code. These modifications act as a dynamic switch that regulates protein function by altering protein localization and activity, as well as interactions with other molecules, lipids, glycans, and nucleic acids. Most PTMs utilize charged or polar groups as the covalent modifications that trigger this switch. Protein S-palmitoylation is one of a group of PTMs that differs from most others in that a hydrophobic, lipid-like group is linked to the substrate during the modification. For S-palmitoylation, usually the fatty acid palmitate is the added modification. As a result, the modification causes cytoplasmic proteins to be first targeted to and then to associate with the cytoplasmic leaflet of the plasma membrane or other organellar membranes. Integral membrane proteins are also S-palmitoylated. However, the role of S-palmitoylation of membrane proteins is more subtle than for cytoplasmic proteins, aiding in the sorting of proteins to specialized lipid domains such as lipid rafts (see Hayashi article below) or the selective trafficking between endomembranes along the secretory pathway (Smotrys and Linder, 2004; Aicart-Ramos et al., 2011).

The most common type of palmitoylation, S-palmitoylation, is reversible, occurs via thioester linkage to cysteine residues, and is catalyzed by palmitoyl acyltransferases (PATs). This is distinct from other forms of protein palmitoylation (e.g., N-palmityolation), which are irreversible and use a different set of palmitoylating enzymes (Linder and Deschenes, 2007; Resh, 2021). PATs are large, multi-pass integral membrane proteins containing a conserved catalytic site positioned near the inner membrane leaflet (Fukata et al., 2004; Greaves and Chamberlain, 2011; Korycka et al., 2012). This conserved region consists of the aspartate-histidine-histidine-cysteine (DHCC) sequence, and thus, PATs are also referred to as zDHHC proteins (Fukata et al., 2004; Greaves and Chamberlain, 2011; Korycka et al., 2012). Considering S-palmitoylation is typically a dynamic and reversible process, both the palmitoylation and depalmitoylation processes act to regulate modified proteins. Depalmitolyation is mediated by cytoplasmic thioesterases which are still being characterized (Lin and Conibear, 2015a,b; Won et al., 2018; Koster and Yoshii, 2019).

The importance of S-palmitoylation is evident, since this modification occurs on ~11% of the human proteome (Blanc et al., 2015). This modification and other PTMs are highly prevalent in the proteome (Kang et al., 2008; Sanders et al., 2015), as their dynamic nature coincides with the shuttling of molecules (or groups of molecules) to and from synaptic domain being the basis for plasticity. In the nearly 5 years since the first volume of this Research Topic was published (Yoshii and Green, 2020), more progress has been made in characterizing how protein S-palmitoylation influences synaptic form and function, neuronal plasticity, and the brain at the systems level. Much of the most recent work centers on the influence of synaptic activity/plasticity on synaptic protein palmitoylation, building on the seminal findings of Noritake et al. (2009). For instance, several new studies demonstrate activity-dependent changes to synaptic protein palmitoylation, including the enzymes that mediate palmitoylation themselves (Nasseri et al., 2022; Abazari et al., 2023). Similarly, multiple lines of experiments (Shen et al., 2022; Koster et al., 2023) reveal that the palmitoylation of postsynaptic scaffolds, neurotransmitter receptors, and other synaptic molecules are crucial for mediating homeostatic synaptic plasticity (Turrigiano et al., 1998; Turrigiano, 1999). Finally, recent work also solidifies a role for synaptic protein palmitoylation in various forms of plasticity that may underlie learning across different species (Nelson et al., 2020; Nasseri et al., 2022; Seo et al., 2022). In the current Research Topic, several groups have examined how palmitoylation of synaptic proteins affects their precise localization, interactions with other synaptic molecules, and the downstream consequences of dysregulated synaptic palmitoylation.

In a brief article reporting new findings with respect to long-term plasticity at glutamatergic synapses, the Hayashi lab presents new evidence that phosphorylation of the GluA2 subunit of AMPA receptor at tyrosine 876 (GluA2-pTyr876) requires intact lipid rafts. Specifically, Hayashi demonstrates that Tyr876 phosphorylated GluA2 is largely localized to a biochemically isolated lipid raft fraction in primary cortical neurons. Further, disruption of lipid rafts abolished the typical stimulation-dependent phosphorylation of GluA2 at Tyr827. Together, these results suggest a crucial role for palmitoylation of Src member kinases like Fyn in the selective trafficking of GluA2 to lipid rafts, providing a model by which palmitoylation controls the compartmentalization of kinases to regulate synaptic strength.

A study from Barylko et al., examines the palmitoylation-dependent localization of calmodulin kinase-like vesicle-associated (CaMKv), a pseudokinase that is required for the maintenance of dendritic spines and typical synaptic physiology (Liang et al., 2016). The authors demonstrate that CaMKv is indeed palmitoylated at the predicted site (cysteine 5; Collins et al., 2017) and that its palmitoylation is required for localization of CaMKv to the plasma membrane. Intriguingly, the article also shows that CaMKv directly interacts with the immediate early gene, Arc, in a palmitoylation-dependent fashion. These data suggest that the palmitoylation of both Arc and CaMKv are involved in the activity-dependent endo- and exocytosis of AMPA receptors during Hebbian plasticity.

The studies from Chen et al., and the Yoshii lab group both provide novel insight into how palmitoylation of the A-kinase anchoring protein 5 (Akap5) influences its role as a postsynaptic scaffold. First, a detailed light and electron microscopic study of Akap5 nanoscale organization demonstrates that N-terminal palmitoylation of Akap5 is necessary for it adapting a vertical (i.e., extended) orientation relative to the postsynaptic membrane, similarly to PSD-95 (Jeyifous et al., 2016). Palmitoylated Akap5 is also more likely to associate with the postsynaptic density and synaptic endosomes, while palmitoylation had a lesser effect on the extrasynaptic Akap5 pool. Performing the same analyses with a palmitoylation-deficient mutant of Akap5 revealed that concurrent with loss of this characteristic vertical, extended orientation, it is displaced from the postsynaptic density. These findings indicate that palmitoylation of Akap5 underpins its stability in synaptic sub-compartments, like the postsynaptic density.

In a series of experiments, the lab of Dr. Akira Yoshii demonstrates that Akap5 is overly palmitoylated in a model of infantile Batten disease, which occurs due to a mutation in a depalmitoylating enzyme. Building on the evidence from Chen et al. here, as well as previous studies on Akap5 function (Purkey et al., 2018; Sanderson et al., 2018), they demonstrate that the excessive palmitoylation of Akap5 in this disease model correlates with a plasticity-dependent excessive upregulation of the AMPA receptor subunit GluA1. Further scrutiny implied that aberrant Akap5 palmitoylation links synaptic receptor activation to downstream pro-inflammatory signaling through its transcriptional targets, which exacerbated the Batten disease phenotype under these circumstances. Taken together, these studies point to the palmitoylation state of Akap5 as a critical regulator of glutamate receptors and local signaling pathways.

Author contributions

KK: Writing – original draft, Writing – review & editing. WG: Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abazari, D., Wild, A. R., Qiu, T., Dickinson, B. C., and Bamji, S. X. (2023). Activity-dependent post-translational regulation of palmitoylating and depalmitoylating enzymes in the hippocampus. J. Cell Sci. 136:jcs260629. doi: 10.1242/jcs.260629

PubMed Abstract | Crossref Full Text | Google Scholar

Aicart-Ramos, C., Valero, R. A., and Rodriguez-Crespo, I. (2011). Protein palmitoylation and subcellular trafficking. Biochim. Biophys. Acta Biomembr. 1808, 2981–2994. doi: 10.1016/j.bbamem.2011.07.009

PubMed Abstract | Crossref Full Text | Google Scholar

Blanc, M., David, F., Abrami, L., Migliozzi, D., Armand, F., Bürgi, J., et al. (2015). SwissPalm: protein palmitoylation database. F1000Research 4:261. doi: 10.12688/f1000research.6464.1

PubMed Abstract | Crossref Full Text | Google Scholar

Collins, M. O., Woodley, K. T., and Choudhary, J. S. (2017). Global, site-specific analysis of neuronal protein S-acylation. Sci. Rep. 7:4683. doi: 10.1038/s41598-017-04580-1

PubMed Abstract | Crossref Full Text | Google Scholar

Fukata, M., Fukata, Y., Adesnik, H., Nicoll, R. A., and Bredt, D. S. (2004). Identification of PSD-95 palmitoylating enzymes. Neuron 44, 987–996. doi: 10.1016/j.neuron.2004.12.005

PubMed Abstract | Crossref Full Text | Google Scholar

Greaves, J., and Chamberlain, L. H. (2011). DHHC palmitoyl transferases: substrate interactions and (patho)physiology. Trends Biochem. Sci. 36, 245–253. doi: 10.1016/j.tibs.2011.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

Jeyifous, O., Lin, E. I., Chen, X., Antinone, S. E., Mastro, R., Drisdel, R., et al. (2016). Palmitoylation regulates glutamate receptor distributions in postsynaptic densities through control of PSD95 conformation and orientation. Proc. Natl. Acad. Sci. U. S. A. 113, E8482–E8491. doi: 10.1073/pnas.1612963113

PubMed Abstract | Crossref Full Text | Google Scholar

Kang, R., Wan, J., Arstikaitis, P., Takahashi, H., Huang, K., Bailey, A. O., et al. (2008). Neural palmitoyl-proteomics reveals dynamic synaptic palmitoylation. Nature 456, 904–909. doi: 10.1038/nature07605

PubMed Abstract | Crossref Full Text | Google Scholar

Korycka, J., Łach, A., Heger, E., Bogusławska, D. M., Wolny, M., Toporkiewicz, M., et al. (2012). Human DHHC proteins: a spotlight on the hidden player of palmitoylation. Eur. J. Cell Biol. 91, 107–117. doi: 10.1016/j.ejcb.2011.09.013

PubMed Abstract | Crossref Full Text | Google Scholar

Koster, K. P., Flores-Barrera, E., Villarmois, E. A., de la Caballero, A., Tseng, K. Y., and Yoshii, A. (2023). Loss of depalmitoylation disrupts homeostatic plasticity of AMPARs in a mouse model of infantile neuronal ceroid lipofuscinosis. J. Neurosci. 43, 8317–8335. doi: 10.1523/JNEUROSCI.1113-23.2023

PubMed Abstract | Crossref Full Text | Google Scholar

Koster, K. P., and Yoshii, A. (2019). Depalmitoylation by palmitoyl-protein thioesterase 1 in neuronal health and degeneration. Front. Synapt. Neurosci. 11:25. doi: 10.3389/fnsyn.2019.00025

PubMed Abstract | Crossref Full Text | Google Scholar

Liang, Z., Zhan, Y., Shen, Y., Wong, C. C. L., Yates, J. R., Plattner, F., et al. (2016). The pseudokinase CaMKv is required for the activity-dependent maintenance of dendritic spines. Nat. Commun. 7:13282. doi: 10.1038/ncomms13282

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, D. T., and Conibear, E. (2015a). ABHD17 proteins are novel protein depalmitoylases that regulate N-Ras palmitate turnover and subcellular localization. Elife 4:e11306. doi: 10.7554/eLife.11306

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, D. T., and Conibear, E. (2015b). Enzymatic protein depalmitoylation by acyl protein thioesterases. Biochem. Soc. Trans. 43, 193–198. doi: 10.1042/BST20140235

PubMed Abstract | Crossref Full Text | Google Scholar

Linder, M. E., and Deschenes, R. J. (2007). Palmitoylation: policing protein stability and traffic. Nat. Rev. Mol. Cell Biol. 8, 74–84. doi: 10.1038/nrm2084

PubMed Abstract | Crossref Full Text | Google Scholar

Nasseri, G. G., Matin, N., Wild, A. R., Tosefsky, K., Flibotte, S., Stacey, R. G., et al. (2022). Synaptic activity–dependent changes in the hippocampal palmitoylome. Sci. Signal. 15:eadd2519. doi: 10.1126/scisignal.add2519

PubMed Abstract | Crossref Full Text | Google Scholar

Nelson, J. C., Witze, E., Ma, Z., Ciocco, F., Frerotte, A., Randlett, O., et al. (2020). Acute regulation of habituation learning via posttranslational palmitoylation. Curr. Biol. 30, 2729–2738.e4. doi: 10.1016/j.cub.2020.05.016

PubMed Abstract | Crossref Full Text | Google Scholar

Noritake, J., Fukata, Y., Iwanaga, T., Hosomi, N., Tsutsumi, R., Matsuda, N., et al. (2009). Mobile DHHC palmitoylating enzyme mediates activity-sensitive synaptic targeting of PSD-95. J. Cell Biol. 186, 147–160. doi: 10.1083/jcb.200903101

PubMed Abstract | Crossref Full Text | Google Scholar

Purkey, A. M., Woolfrey, K. M., Crosby, K. C., Stich, D. G., Chick, W. S., Aoto, J., et al. (2018). AKAP150 palmitoylation regulates synaptic incorporation of Ca2+-permeable AMPA receptors to control LTP. Cell Rep. 25, 974–987.e4. doi: 10.1016/j.celrep.2018.09.085

PubMed Abstract | Crossref Full Text | Google Scholar

Resh, M. D. (2021). Palmitoylation of Hedgehog proteins by Hedgehog acyltransferase: roles in signalling and disease. Open Biol. 11:200414. doi: 10.1098/rsob.200414

PubMed Abstract | Crossref Full Text | Google Scholar

Sanders, S. S., Martin, D. D., Butland, S. L., Lavallée-Adam, M., Calzolari, D., Kay, C., et al. (2015). Curation of the mammalian palmitoylome indicates a pivotal role for palmitoylation in diseases and disorders of the nervous system and cancers. PLoS Comput. Biol. 11:e1004405. doi: 10.1371/journal.pcbi.1004405

PubMed Abstract | Crossref Full Text | Google Scholar

Sanderson, J. L., Scott, J. D., and Dell'Acqua, M. L. (2018). Control of homeostatic synaptic plasticity by AKAP-anchored kinase and phosphatase regulation of Ca 2+ -permeable AMPA receptors. J. Neurosci. 38, 2863–2876. doi: 10.1523/JNEUROSCI.2362-17.2018

PubMed Abstract | Crossref Full Text | Google Scholar

Seo, J., Hwang, H., Choi, Y., Jung, S., Hong, J.-H., Yoon, B.-J., et al. (2022). Myristoylation-dependent palmitoylation of cyclin Y modulates long-term potentiation and spatial learning. Prog. Neurobiol. 218:102349. doi: 10.1016/j.pneurobio.2022.102349

PubMed Abstract | Crossref Full Text | Google Scholar

Shen, Z.-C., Xia, Z.-X., Liu, J.-M., Zheng, J.-Y., Luo, Y.-F., Yang, H., et al. (2022). APT1-mediated depalmitoylation regulates hippocampal synaptic plasticity. J. Neurosci. 42, 2662–2677. doi: 10.1523/JNEUROSCI.1741-21.2022

PubMed Abstract | Crossref Full Text | Google Scholar

Smotrys, J. E., and Linder, M. E. (2004). Palmitoylation of intracellular signaling proteins: regulation and function. Annu. Rev. Biochem. 73, 559–587. doi: 10.1146/annurev.biochem.73.011303.073954

PubMed Abstract | Crossref Full Text | Google Scholar

Turrigiano, G., Leslie, K., Desai, N., Rutherford, L., and Nelson, S. (1998). Activity-dependent scaling of quantal amplitude in neocortical neurons. Nature 391, 892–896. doi: 10.1038/36103

PubMed Abstract | Crossref Full Text | Google Scholar

Turrigiano, G. G. (1999). Homeostatic plasticity in neuronal networks: the more things change, the more they stay the same. Trends Neurosci. 22, 221–227. doi: 10.1016/S0166-2236(98)01341-1

PubMed Abstract | Crossref Full Text | Google Scholar

Won, S. J., Kit, M. C. S., and Martin, B. R. (2018). Protein depalmitoylases. Crit. Rev. Biochem. Mol. Biol. 53, 83–98. doi: 10.1080/10409238.2017.1409191

PubMed Abstract | Crossref Full Text | Google Scholar

Yoshii, A., and Green, W. N. (2020). Editorial: role of protein palmitoylation in synaptic plasticity and neuronal differentiation. Front. Synapt. Neurosci. 12:27. doi: 10.3389/fnsyn.2020.00027

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: palmitoylation, depalmitoylation, synaptic transmission, synaptic plasticity, post-translational modification

Citation: Koster KP and Green WN (2024) Editorial: Role of protein palmitoylation in synaptic plasticity and neuronal differentiation, volume II. Front. Synaptic Neurosci. 16:1473989. doi: 10.3389/fnsyn.2024.1473989

Received: 31 July 2024; Accepted: 26 August 2024;
Published: 10 September 2024.

Edited and reviewed by: P. Jesper Sjöström, McGill University, Canada

Copyright © 2024 Koster and Green. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Kevin P. Koster, kpkoster@uchicago.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.