Dysfunctional S1P/S1PR1 signaling in the dentate gyrus drives vulnerability of chronic pain-related memory impairment

  1. Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
  2. Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
  3. NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
  4. School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
  5. Faculty of Health Sciences, University of Macau, Taipa, Macau, China
  6. Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, China

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Mychael Lourenco
    Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
  • Senior Editor
    Lu Chen
    Stanford University, Stanford, United States of America

Reviewer #1 (Public Review):

This work from Cui, Pan, Fan, et al explores memory impairment in chronic pain mouse models, a topic of great interest in the neurobiology field. In particular, the work starts from a very interesting observation, that WT mice can be divided into susceptible and unsusceptible to memory impairment upon modelling chronic pain with CCI. This observation represents the basis of the work where the authors identify the sphingosine receptor S1PR1 as down-regulated in the dentate gyrus of susceptible animals and demonstrate through an elegant range of experiments involving AAV-mediated knockdown or overexpression of S1PR1 that this receptor is involved in the memory impairment observed with chronic pain. Importantly for translational purposes, they also show that activation of S1PR1 through a pharmacological paradigm is able to rescue the memory impairment phenotype.

The authors also link these defects to reduced dendritic branching and a reduced number of mature excitatory synapses in the DG to the memory phenotype.

They then proceed to explore possible mechanisms downstream of S1PR1 that could explain this reduction in dendritic spines. They identify integrin α2 as an interactor of S1PR1 and show a reduction in several proteins involved in actin dynamic, which is crucial for dendritic spine formation and plasticity.

They thus hypothesize that the interaction between S1PR1 and Integrin α2 is fundamental for the activation of Rac1 and Cdc42 and consequently for the polymerisation of actin; a reduction in this pathway upon chronic pain would thus lead to impaired actin polymerisation, synapse formation, and thus impaired memory.

The work is of great interest and the experiments are of very good quality with results of great importance. I have however some concerns. The main concern I have relates to the last part of the work, namely Figures 8 and 9, which I feel are not at the same level as the results presented in the previous 7 Figures, which are instead outstanding.

In particular:

- In Figure 8, given the reduction in all the proteins tested, the authors need to check some additional proteins as controls. One good candidate could be RhoA, considering the authors say it is activated by S1PR2 and not by S1PR1;

- In addition to the previous point, could the authors also show that the number of neurons is not grossly different between susceptible and unsusceptible mice? This could be done by simply staining for NeuN or performing a western blot for a neuronal-specific protein (e.g. Map2 or beta3-tubulin);

- In Figure 8, the authors should also evaluate the levels of activated RAC1 and activated Cdc42, which are much more important than just basal levels of the proteins to infer an effect on actin dynamics. This is possible through kits that use specific adaptors to pulldown GTP-Rac1 and GTP-Cdc42;

- In Figure 9C, the experiment is performed in an immortalised cell line. I feel this needs to be performed at least in primary hippocampal neurons;

- In Figure 9D, the authors use a Yeast two-hybrid system to demonstrate the interaction between S1PR1 and Integrin α2. However, as the yeast two-hybrid system is based on the proximity of the GAL4 activating domain and the GAL4 binding domain, which are used to activate the transcription of reporter genes, the system is not often used when probing the interaction between transmembrane proteins. Could the authors use other transmembrane proteins as negative controls?;

- In Figure 9E, the immunoblot is very unconvincing. The bands in the inputs are very weak for both ITGA2 and S1PR1, the authors do not show the enrichment of S1PR1 upon its immunoprecipitation and the band for ITGA2 in the IP fraction has a weird appearance. Were these experiments performed on DG lysates only? If so, I suggest the authors repeat the experiment using the whole brain (or at least the whole hippocampus) so as to have more starting material. Alternatively, if this doesn't work, or in addition, they could also perform the immunoprecipitation in heterologous cells overexpressing the two proteins;

- About the point above, even if the results were convincing, the authors can't say that they demonstrate an interaction in vivo. In co-IP experiments, the interaction is much more likely to occur in the lysate during the incubation period rather than being conserved from the in vivo state. These co-IPs demonstrate the ability of proteins to interact, not necessarily that they do it in vivo. If the authors wanted to demonstrate this, they could perform a Proximity ligation assay in primary hippocampal neurons, using antibodies against S1PR1 and ITGA2.

- In Figure 9H, could the authors increase the N to see if shItga2 causes further KD in the CCI?

- To conclusively demonstrate that S1PR1 and ITGA2 participate in the same pathway, they could show that knocking down the two proteins at the same time does not have additive effects on behavioral tests compared to the knockdown of each one of them in isolation.

Other major concerns:

- Supplementary Figure 5: the image showing colocalisation between S1PR1 and CamKII is not very convincing. Is the S1PR1 antibody validated on Knockout or knockdown in immunostaining?;

- It would be interesting to check S1PR2 levels as a control in CCI-chronic animals;

- Figure 1: I am a bit concerned about the Ns in these experiments. In the chronic pain experiments, the N for Sham is around 8 whereas is around 20 for CCI animals. Although I understand higher numbers are necessary to see the susceptible and unsusceptible populations, I feel that then the same number of Sham animals should be used;

- Figures 1E and 1G have much higher Ns than the other panels. Why is that? If they have performed this high number of animals why not show them in all panels?;

- In the experiments where viral injection is performed, the authors should show a zoomed-out image of the brain to show the precision of the injection and how spread the expression of the different viruses was;

- The authors should check if there is brain inflammation in CCI chronic animals. This would be interesting to explain if this could be the trigger for the effects seen in neurons. In particular, the authors should check astrocytes and microglia. This is of interest also because the pathways altered in Figure 8A are related to viral infection;

- If the previous point shows increased brain inflammation, it would be interesting for the authors to check whether a prolonged anti-inflammatory treatment in CCI animals administered before the insurgence of memory impairment could stop it from happening;

- In addition, the authors should speculate on what could be the signal that can induce these molecular changes starting from the site of injury;

- Also, as the animals are all WT, the authors should speculate on what could render some animals prone to have memory impairments and others resistant.

Reviewer #2 (Public Review):

Summary:

The study investigates the molecular mechanisms underlying chronic pain-related memory impairment by focusing on S1P/S1PR1 signaling in the dentate gyrus (DG) of the hippocampus. Through behavioural tests (Y-maze and Morris water maze) and RNA-seq analysis, the researchers segregated chronic pain mice into memory impairment-susceptible and -unsusceptible subpopulations. They discovered that S1P/S1PR1 signaling is crucial for determining susceptibility to memory impairment, with decreased S1PR1 expression linked to structural plasticity changes and memory deficits.

Knockdown of S1PR1 in the DG induced a susceptible phenotype, while overexpression or pharmacological activation of S1PR1 promoted resistance to memory impairment and restored normal synaptic structure. The study identifies actin cytoskeleton-related pathways, including ITGA2 and its downstream Rac1/Cdc42 signaling, as key mediators of S1PR1's effects, offering new insights and potential therapeutic targets for chronic pain-related cognitive dysfunction.

This manuscript consists of a comprehensive investigation and significant findings. The study provides novel insights into the molecular mechanisms of chronic pain-related memory impairment, highlighting the critical role of S1P/S1PR1 signaling in the hippocampal dentate gyrus. The clear identification of S1P/S1PR1 as a potential therapeutic target offers promising avenues for future research and treatment strategies. The manuscript is well-structured, methodologically sound, and presents valuable contributions to the field.

Strengths:

(1) The manuscript is well-structured and written in clear, concise language. The flow of information is logical and easy to follow.

(2) The segregation of mice into memory impairment-susceptible and -unsusceptible subpopulations is innovative and well-justified. The statistical analyses are robust and appropriate for the data.

(3) The detailed examination of S1PR1 expression and its impact on synaptic plasticity and actin cytoskeleton reorganization is impressive. The findings are significant and contribute to the understanding of chronic pain-related memory impairment.

Weaknesses:

(1) Results: While the results are comprehensive, some sections are data-heavy and could be more reader-friendly with summarized key points before diving into detailed data.

(2) Discussion: There is a need for a more balanced discussion regarding the limitations of the study. For example, addressing potential biases in the animal model or limitations in the generalizability of the findings to humans would strengthen the discussion. Also, providing specific suggestions for follow-up studies would be beneficial.

(3) Conclusion: The conclusion, while concise, could better highlight the study's broader impact on the field and potential clinical implications.

Reviewer #3 (Public Review):

Summary of the Authors' Objectives:

The authors aimed to delineate the role of S1P/S1PR1 signaling in the dentate gyrus in the context of memory impairment associated with chronic pain. They sought to understand the molecular mechanisms contributing to the variability in memory impairment susceptibility and to identify potential therapeutic targets.

Major Strengths and Weaknesses of the Study:

The study is methodologically robust, employing a combination of RNA-seq analysis, viral-mediated gene manipulation, and pharmacological interventions to investigate the S1P/S1PR1 pathway. The use of both knockdown and overexpression approaches to modulate S1PR1 levels provides compelling evidence for its role in memory impairment. The research also benefits from a comprehensive assessment of behavioral changes associated with chronic pain.

However, the study has some weaknesses. The categorization of mice into 'susceptible' and 'unsusceptible' groups based on memory performance requires further validation. Additionally, the reliance on a single animal model may limit the generalizability of the findings. The study could also benefit from a more detailed exploration of the impact of different types of pain on memory impairment.

Assessment of the Authors' Achievements:

The authors successfully identified S1P/S1PR1 signaling as a key factor in chronic pain-related memory impairment and demonstrated its potential as a therapeutic target. The findings are supported by rigorous experimental evidence, including biochemical, histological, and behavioral data. However, the study's impact could be enhanced by further exploration of the molecular pathways downstream of S1PR1 and by assessing the long-term effects of S1PR1 manipulation.

Impact on the Field and Utility to the Community:

This study is likely to have a significant impact on pain research by providing a novel perspective on the mechanisms underlying memory impairment in chronic pain conditions. The identification of the S1P/S1PR1 pathway as a potential therapeutic target could guide the development of new treatments.

Additional Context for Readers:

The study's approach to categorizing susceptibility to memory impairment could inspire new methods for stratifying patient populations in clinical settings.

Recommendations:

(1) A more detailed explanation of the k-means clustering algorithm and its application in categorizing mice should be provided.

(2) The discussion on the potential influence of different pain types or sensitivities on memory impairment should be expanded.

(3) The protocol for behavioral testing should be clarified and the potential for learning or stress effects should be addressed.

(4) Conduct additional behavioral assays for other molecular targets implicated in the study.

(5) The effective drug thresholds and potential non-specific effects of pharmacological interventions should be discussed in more detail.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation