Skip to main content

Main menu

  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
  • EDITORIAL BOARD
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
  • SUBSCRIBE

User menu

  • Log in
  • My Cart

Search

  • Advanced search
Journal of Neuroscience
  • Log in
  • My Cart
Journal of Neuroscience

Advanced Search

Submit a Manuscript
  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
  • EDITORIAL BOARD
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
  • SUBSCRIBE
PreviousNext
Journal Club

Effects of Astroglia on Motor Neurons in Spinal Muscular Atrophy

Bert M. Verheijen
Journal of Neuroscience 6 September 2017, 37 (36) 8578-8580; DOI: https://doi.org/10.1523/JNEUROSCI.1578-17.2017
Bert M. Verheijen
Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Bert M. Verheijen
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Hereditary proximal spinal muscular atrophy (SMA), a severe neuromuscular disorder and a leading genetic cause of infant death, is characterized by loss of motor neurons in the ventral horn of the spinal cord, resulting in progressive muscle atrophy and weakness (Lunn and Wang, 2008). SMA is an autosomal recessive disease that is caused by homozygous disruptions of the survival motor neuron 1 (SMN1) gene, resulting in reduced levels of SMN protein (Lefebvre et al., 1997). Although humans have a second, highly homologous copy of the SMN gene, named SMN2, it undergoes alternative splicing that truncates the transcript and therefore only produces small amounts of full-length SMN protein. Nonetheless, SMN2 copy number variation in patients is a major modifier of disease severity, because the number of copies directly influences SMN protein levels.

Although the genetic underpinnings of SMA have been known for several decades, it is not clear how loss of SMN gives rise to SMA (Monani, 2005; Burghes and Beattie, 2009). Previous work has indicated that SMN is involved in several cellular processes, however, including pre-mRNA splicing, transcription termination, RNA trafficking, local translation regulation and stress granule formation (Monani, 2005; Burghes and Beattie, 2009; Singh et al., 2017). Many studies on SMA focus exclusively on the perturbation of these processes in motor neurons as a result of SMN deficiency. However, accumulating evidence suggests that other cell types, including non-neuronal cells like neuroglia, also play important roles in SMA (Hamilton and Gillingwater, 2013). A convincing argument for the involvement of non-motor neuron cells in SMA is the finding that only systemic restoration of SMN levels in SMA mouse models provides long-term rescue of the phenotype: increasing SMN in neurons alone is less effective (Hua et al., 2011; Martinez et al., 2012). Moreover, Schwann cells, the predominant glial cells of the peripheral nervous system that produce myelin and regulate neuromuscular junction formation and maintenance, show intrinsic defects in transgenic mouse models for SMA (Hunter et al., 2014, 2016). Therefore, insight into the contribution of non-neuronal cells is crucial for a better understanding of SMA pathology, and it may lead to the identification of novel therapeutic targets.

Astrocytes have also been found to contribute to SMA. These cells carry out numerous vital functions in the nervous system. For example, they influence synaptic communication and they support neuronal development and survival by secreting multiple beneficial factors (e.g., growth factors). Astrocytes are increasingly recognized as key players in several neurodegenerative diseases, including motor neuron disease (Pekny et al., 2016). SMN-deficient astrocytes show marked alterations and impairments, such as increased expression of glial fibrillary acidic protein and shorter process length (which are indicative of reactive astrocytosis), abnormal calcium homeostasis, reduced neurotrophin production, and diminished support of motor neuron synapse formation in vitro (McGivern et al., 2013; Zhou et al., 2016). Notably, a selective increase of SMN in astrocytes increases life span in SMA mice, although it does not prevent motor neuron death (Rindt et al., 2015). How astrocytes mediate these effects in SMA remains unknown.

In a recent study published in The Journal of Neuroscience, Martin et al. (2017) revealed a novel aspect of astrocyte dysfunction in SMA. Through the use of astrocyte-conditioned medium (ACM), i.e., medium that had been incubated with SMN-deficient murine astrocytes, they observed motor neuron defects in culture and identified a potential role for astrocyte-secreted monocyte chemoattractant protein 1 (CCL2/MCP1) in causing these defects.

The authors isolated primary astrocytes from spinal cord of both wild-type (WT) and SMA neonatal mice. These astrocytes were cultured for 1 week, and medium that had been on these cultures was collected every 48 h. Collected ACM was then added to cultures of isolated primary mouse motor neurons or motor neurons derived from human induced pluripotent stem cells (hiPSCs). Compared with neurons cultured in WT ACM, those grown in medium from SMA astrocytes had significantly shorter neurites and lower expression of motor neuron-specific expression markers, indicating motor neuron loss (Martin et al., 2017, their Figs. 1, 2A,B). hiPSC-motor neurons derived from SMA patient fibroblasts showed similar neurite aberrations when cultured in SMA ACM (Martin et al., 2017, their Fig. 2C,D). This indicated that SMA ACM negatively affects both WT and SMA motor neuron health in vitro.

To identify the factors present or lacking in the SMA ACM that are responsible for these detrimental effects on motor neurons, Martin et al. (2017) compared the expression of 32 chemokines in WT and SMA ACM. ELISAs were subsequently performed to measure protein concentration of select chemokines. Of course, the astrocyte secretome contains many more factors than these 32 chemokines, pointing out a drawback to this experimental approach. Nevertheless, ELISA showed a substantial decrease of MCP1 protein levels in SMA ACM compared with WT ACM (Martin et al., 2017, their Fig. 3).

MCP1 is a well-studied chemotactic protein that has been previously found to improve neuron survival and promote neuronal activity and neurite outgrowth in vitro. Both motor neurons and astrocytes have been shown to express CCR2, a receptor for MCP1 (see references in Martin et al., 2017). Martin et al. (2017) measured MCP1 mRNA levels in WT and SMA mouse spinal cords of different ages (Martin et al., 2017, their Fig. 4), and found reduced MCP1 levels in spinal cords of young mice with SMA. To determine whether MCP1 affects neuronal survival and neurite outgrowth in culture, primary mouse motor neurons were supplemented with recombinant MCP1. The addition of exogenous MCP1 stimulated neurite outgrowth and increased the expression of a motor neuron marker (Martin et al., 2017, their Fig. 5). Similarly, MCP1 supplementation increased neurite outgrowth in WT and SMA hiPSC-derived motor neurons (Martin et al., 2017, their Fig. 6). Importantly, the addition of exogenous MCP1 to astrocytes alone did not influence the effects of ACM on neurons, demonstrating that the effect on motor neurons is direct and is not due to autocrine signaling on astrocytes. Conversely, addition of an anti-MCP1 neutralizing antibody to WT ACM impaired neurite outgrowth and reduced the expression of motor neuron markers in primary mouse motor neurons as well as in WT and SMA hiPSC motor neurons (Martin et al., 2017, their Figs. 7, 8).

Overall, these results suggest that abnormal levels of glia-secreted factors, including the chemokine MCP1, in SMA contribute to defects in cultured motor neurons (Fig. 1).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Abnormal motor neuron–glia interactions in SMA. Astrocytes secrete factors to support neuron development and function. Reduced levels of secreted factors derived from cultures of murine astrocytes, specifically MCP1, induced motor neuron defects in culture. Glia and glia-secreted molecules could act as important drivers of SMA pathology. Astrocyte image based on original image from Cancer Research UK.

Martin et al. (2017) propose that the restoration of deficient astrocyte-secreted factors be explored as a therapeutic approach. Perhaps such an approach could complement therapies that aim to increase SMN protein levels (e.g., via gene therapy and SMN2-targeting strategies; Foust et al., 2010; Corey, 2017; d'Ydewalle et al., 2017). However, several questions should be addressed before taking this step.

First, no data on the purity of the astrocyte cultures is provided. Therefore, it is possible that other cells, including immunoregulatory cells like microglia, were present in the astrocyte cultures that were used. Reduced numbers or secretion of MCP1 by such cells in the SMA condition may have contributed to the observed defects. Based on previous expression data, it seems likely that microglia are an important source of MCP1 (Orre et al., 2014; Zhang et al., 2014, 2016). Future work could address this possibility by measuring the expression of microglia marker Iba-1 in cultures or by purifying the astrocytes via fluorescence-activated cell sorting. If future therapies target particular cell types, it will be important to accurately identify the cells and mechanisms that mediate specific SMA-related defects. Yet, supplementing motor neurons with MCP1 remains a potential therapeutic option, regardless of the cellular source.

Another unresolved question is how MCP1 achieves its advantageous downstream effects on motor neurons. It has been previously shown that MCP1 can be protective against HIV-1 transactivator protein (Tat)-induced neurotoxicity, in a CCR2-dependent manner (Yao et al., 2009). Removal or blocking of CCR2 on motor neurons would reveal whether the effect of MCP1 is mediated by CCR2 signaling or via other pathways. Through such pathways, MCP1 might, for example, inhibit apoptosis or prevent excitotoxicity in neurons (Madrigal et al., 2009). Dissecting the mechanisms in more detail could result in novel therapeutic targets and enhanced therapeutic efficacy.

Finally, as pointed out by Martin et al. (2017), modulating chemokine levels might not be a viable approach in patients because of the proinflammatory properties of these molecules. In fact, there is evidence that elevation of MCP1 levels and gliosis occur as SMA progresses (Rindt et al., 2015). Astrocytic loss-of-function during early development might shift toward a toxic gain-of-function in later stages of the disease, narrowing the therapeutic window of opportunity and the range of efficacy in patients. It is worth noting that the role of neuroinflammation and microglia, major immune effector cells in the nervous system, has been largely unexplored in SMA (Rindt et al., 2015). It is possible, for example, that abnormal synaptic pruning by microglia contributes to the motor neuron deafferentation observed in SMA. Interactions of astrocyte-secreted factors with other cells, including Schwann cells, microglia, and other astrocytes, should be investigated before seriously considering the use of such factors as a therapy.

In summary, glia-secreted factors appear to be an important contributor to motor neuron defects in SMA. Future studies should aim to investigate the variety of secreted factors and the roles of these molecules in more detail. These insights might also be relevant to other neurological disorders. Upcoming studies will benefit from advances in the generation of different types of hiPSC-derived glial cells and gene-editing techniques to accurately mimic patient cells in a nonxenogeneic setup. Nonetheless, any results generated in vitro must ultimately be validated in in vivo experimental model systems for SMA. It would be important to confirm whether the restoration of glia-secreted factors, such as MCP1, ameliorates disease progression in SMA mice before moving on to develop therapies.

Footnotes

  • Editor's Note: These short reviews of recent JNeurosci articles, written exclusively by students or postdoctoral fellows, summarize the important findings of the paper and provide additional insight and commentary. If the authors of the highlighted article have written a response to the Journal Club, the response can be found by viewing the Journal Club at www.jneurosci.org. For more information on the format, review process, and purpose of Journal Club articles, please see http://jneurosci.org/content/preparing-manuscript#journalclub.

  • The author declares no competing financial interests.

  • Correspondence should be addressed to Bert M. Verheijen, Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG, Utrecht, The Netherlands. l.m.verheijen-3{at}umcutrecht.nl

References

  1. ↵
    1. Burghes AH,
    2. Beattie CE
    (2009) Spinal muscular atrophy: why do low levels of survival motor neuron protein make motor neurons sick? Nat Rev Neurosci 10:597–609. doi:10.1038/nrn2670 pmid:19584893
    OpenUrlCrossRefPubMed
  2. ↵
    1. Corey DR
    (2017) Nusinersen, an antisense oligonucleotide drug for spinal muscular atrophy. Nat Neurosci 20:497–499. doi:10.1038/nn.4508 pmid:28192393
    OpenUrlCrossRefPubMed
  3. ↵
    1. d'Ydewalle C,
    2. Ramos DM,
    3. Pyles NJ,
    4. Ng SY,
    5. Gorz M,
    6. Pilato CM,
    7. Ling K,
    8. Kong L,
    9. Ward AJ,
    10. Rubin LL,
    11. Rigo F,
    12. Bennett CF,
    13. Sumner CJ
    (2017) The antisense transcript SMN-AS1 regulates SMN expression and is a novel therapeutic target for spinal muscular atrophy. Neuron 93:66–79. doi:10.1016/j.neuron.2016.11.033 pmid:28017471
    OpenUrlCrossRefPubMed
  4. ↵
    1. Foust KD,
    2. Wang X,
    3. McGovern VL,
    4. Braun L,
    5. Bevan AK,
    6. Haidet AM,
    7. Le TT,
    8. Morales PR,
    9. Rich MM,
    10. Burghes AH,
    11. Kaspar BK
    (2010) Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat Biotechnol 28:271–274. doi:10.1038/nbt.1610 pmid:20190738
    OpenUrlCrossRefPubMed
  5. ↵
    1. Hamilton G,
    2. Gillingwater TH
    (2013) Spinal muscular atrophy: going beyond the motor neuron. Trends Mol Med 19:40–50. doi:10.1016/j.molmed.2012.11.002 pmid:23228902
    OpenUrlCrossRefPubMed
  6. ↵
    1. Hua Y,
    2. Sahashi K,
    3. Rigo F,
    4. Hung G,
    5. Horev G,
    6. Bennett CF,
    7. Krainer AR
    (2011) Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model. Nature 478:123–126. doi:10.1038/nature10485 pmid:21979052
    OpenUrlCrossRefPubMed
  7. ↵
    1. Hunter G,
    2. Aghamaleky Sarvestany A,
    3. Roche SL,
    4. Symes RC,
    5. Gillingwater TH
    (2014) SMN-dependent intrinsic defects in Schwann cells in mouse models of spinal muscular atrophy. Hum Mol Genet 23:2235–2250. doi:10.1093/hmg/ddt612 pmid:24301677
    OpenUrlCrossRefPubMed
  8. ↵
    1. Hunter G,
    2. Powis RA,
    3. Jones RA,
    4. Groen EJ,
    5. Shorrock HK,
    6. Lane FM,
    7. Zheng Y,
    8. Sherman DL,
    9. Brophy PJ,
    10. Gillingwater TH
    (2016) Restoration of SMN in Schwann cells reverses myelination defects and improves neuromuscular function in spinal muscular atrophy. Hum Mol Genet 25:2853–2861. doi:10.1093/hmg/ddw141 pmid:27170316
    OpenUrlCrossRefPubMed
  9. ↵
    1. Lefebvre S,
    2. Burlet P,
    3. Liu Q,
    4. Bertrandy S,
    5. Clermont O,
    6. Munnich A,
    7. Dreyfuss G,
    8. Melki J
    (1997) Correlation between severity and SMN protein level in spinal muscular atrophy. Nat Genet 16:265–269. doi:10.1038/ng0797-265 pmid:9207792
    OpenUrlCrossRefPubMed
  10. ↵
    1. Lunn MR,
    2. Wang CH
    (2008) Spinal muscular atrophy. Lancet 371:2120–2133. doi:10.1016/S0140-6736(08)60921-6 pmid:18572081
    OpenUrlCrossRefPubMed
  11. ↵
    1. Madrigal JL,
    2. Leza JC,
    3. Polak P,
    4. Kalinin S,
    5. Feinstein DL
    (2009) Astrocyte-derived MCP-1 mediates neuroprotective effects of noradrenaline. J Neurosci 29:263–267. doi:10.1523/JNEUROSCI.4926-08.2009 pmid:19129402
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Martin JE,
    2. Nguyen TT,
    3. Grunseich C,
    4. Nofziger JH,
    5. Lee PR,
    6. Fields D,
    7. Fischbeck KH,
    8. Foran E
    (2017) Decreased motor neuron support by SMA astrocytes due to diminished MCP1 secretion. J Neurosci 37:5309–5318. doi:10.1523/JNEUROSCI.3472-16.2017 pmid:28450545
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Martinez TL,
    2. Kong L,
    3. Wang X,
    4. Osborne MA,
    5. Crowder ME,
    6. Van Meerbeke JP,
    7. Xu X,
    8. Davis C,
    9. Wooley J,
    10. Goldhamer DJ,
    11. Lutz CM,
    12. Rich MM,
    13. Sumner CJ
    (2012) Survival motor neuron protein in motor neurons determines synaptic integrity in spinal muscular atrophy. J Neurosci 32:8703–8715. doi:10.1523/JNEUROSCI.0204-12.2012 pmid:22723710
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. McGivern JV,
    2. Patitucci TN,
    3. Nord JA,
    4. Barabas MA,
    5. Stucky CL,
    6. Ebert AD
    (2013) Spinal muscular atrophy astrocytes exhibit abnormal calcium regulation and reduced growth factor production. Glia 61:1418–1428. doi:10.1002/glia.22522 pmid:23839956
    OpenUrlCrossRefPubMed
  15. ↵
    1. Monani UR
    (2005) Spinal muscular atrophy: a deficiency in a ubiquitous protein; a motor neuron-specific disease. Neuron 48:885–896. doi:10.1016/j.neuron.2005.12.001 pmid:16364894
    OpenUrlCrossRefPubMed
  16. ↵
    1. Orre M,
    2. Kamphuis W,
    3. Osborn LM,
    4. Melief J,
    5. Kooijman L,
    6. Huitinga I,
    7. Klooster J,
    8. Bossers K,
    9. Hol EM
    (2014) Acute isolation and transcriptome characterization of cortical astrocytes and microglia from young and aged mice. Neurobiol Aging 35:1–14. doi:10.1016/j.neurobiolaging.2013.07.008 pmid:23954174
    OpenUrlCrossRefPubMed
  17. ↵
    1. Pekny M,
    2. Pekna M,
    3. Messing A,
    4. Steinhäuser C,
    5. Lee JM,
    6. Parpura V,
    7. Hol EM,
    8. Sofroniew MV,
    9. Verkhratsky A
    (2016) Astrocytes: a central element in neurological diseases. Acta Neuropathol 131:323–345. doi:10.1007/s00401-015-1513-1 pmid:26671410
    OpenUrlCrossRefPubMed
  18. ↵
    1. Rindt H,
    2. Feng Z,
    3. Mazzasette C,
    4. Glascock JJ,
    5. Valdivia D,
    6. Pyles N,
    7. Crawford TO,
    8. Swoboda KJ,
    9. Patitucci TN,
    10. Ebert AD,
    11. Sumner CJ,
    12. Ko CP,
    13. Lorson CL
    (2015) Astrocytes influence the severity of spinal muscular atrophy. Hum Mol Genet 24:4094–4102. doi:10.1093/hmg/ddv148 pmid:25911676
    OpenUrlCrossRefPubMed
  19. ↵
    1. Singh RN,
    2. Howell MD,
    3. Ottesen EW,
    4. Singh NN
    (2017) Diverse role of survival motor neuron protein. Biochim Biophys Acta 1860:299–315. doi:10.1016/j.bbagrm.2016.12.008 pmid:28095296
    OpenUrlCrossRefPubMed
  20. ↵
    1. Yao H,
    2. Peng F,
    3. Dhillon N,
    4. Callen S,
    5. Bokhari S,
    6. Stehno-Bittel L,
    7. Ahmad SO,
    8. Wang JQ,
    9. Buch S
    (2009) Involvement of TRPC channels in CCL2-mediated neuroprotection against tat toxicity. J Neurosci 29:1657–1669. doi:10.1523/JNEUROSCI.2781-08.2009 pmid:19211873
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Zhang Y,
    2. Chen K,
    3. Sloan SA,
    4. Bennett ML,
    5. Scholze AR,
    6. O'Keeffe S,
    7. Phatnani HP,
    8. Guarnieri P,
    9. Caneda C,
    10. Ruderisch N,
    11. Deng S,
    12. Liddelow SA,
    13. Zhang C,
    14. Daneman R,
    15. Maniatis T,
    16. Barres BA,
    17. Wu JQ
    (2014) An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34:11929–11947. doi:10.1523/JNEUROSCI.1860-14.2014 pmid:25186741
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Zhang Y,
    2. Sloan SA,
    3. Clarke LE,
    4. Caneda C,
    5. Plaza CA,
    6. Blumenthal PD,
    7. Vogel H,
    8. Steinberg GK,
    9. Edwards MS,
    10. Li G,
    11. Duncan JA 3rd.,
    12. Cheshier SH,
    13. Shuer LM,
    14. Chang EF,
    15. Grant GA,
    16. Gephart MG,
    17. Barres BA
    (2016) Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89:37–53. doi:10.1016/j.neuron.2015.11.013 pmid:26687838
    OpenUrlCrossRefPubMed
  23. ↵
    1. Zhou C,
    2. Feng Z,
    3. Ko CP
    (2016) Defects in motoneuron-astrocyte interactions in spinal muscular atrophy. J Neurosci 36:2543–2553. doi:10.1523/JNEUROSCI.3534-15.2016 pmid:26911699
    OpenUrlAbstract/FREE Full Text
Back to top

In this issue

The Journal of Neuroscience: 37 (36)
Journal of Neuroscience
Vol. 37, Issue 36
6 Sep 2017
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Advertising (PDF)
  • Ed Board (PDF)
Email

Thank you for sharing this Journal of Neuroscience article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Effects of Astroglia on Motor Neurons in Spinal Muscular Atrophy
(Your Name) has forwarded a page to you from Journal of Neuroscience
(Your Name) thought you would be interested in this article in Journal of Neuroscience.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
View Full Page PDF
Citation Tools
Effects of Astroglia on Motor Neurons in Spinal Muscular Atrophy
Bert M. Verheijen
Journal of Neuroscience 6 September 2017, 37 (36) 8578-8580; DOI: 10.1523/JNEUROSCI.1578-17.2017

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Respond to this article
Request Permissions
Share
Effects of Astroglia on Motor Neurons in Spinal Muscular Atrophy
Bert M. Verheijen
Journal of Neuroscience 6 September 2017, 37 (36) 8578-8580; DOI: 10.1523/JNEUROSCI.1578-17.2017
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Responses to this article

Respond to this article

Jump to comment:

No eLetters have been published for this article.

Related Articles

Cited By...

More in this TOC Section

  • Dissociating Hippocampal and Cortical Contributions to Predictive Processing
  • Uncovering the Hippocampal Mechanisms Underpinning Spatial Learning and Flexible Navigation
  • Generalization and Idiosyncrasy: Two Sides of the Same Brain
Show more Journal Club
  • Home
  • Alerts
  • Visit Society for Neuroscience on Facebook
  • Follow Society for Neuroscience on Twitter
  • Follow Society for Neuroscience on LinkedIn
  • Visit Society for Neuroscience on Youtube
  • Follow our RSS feeds

Content

  • Early Release
  • Current Issue
  • Issue Archive
  • Collections

Information

  • For Authors
  • For Advertisers
  • For the Media
  • For Subscribers

About

  • About the Journal
  • Editorial Board
  • Privacy Policy
  • Contact
(JNeurosci logo)
(SfN logo)

Copyright © 2023 by the Society for Neuroscience.
JNeurosci Online ISSN: 1529-2401

The ideas and opinions expressed in JNeurosci do not necessarily reflect those of SfN or the JNeurosci Editorial Board. Publication of an advertisement or other product mention in JNeurosci should not be construed as an endorsement of the manufacturer’s claims. SfN does not assume any responsibility for any injury and/or damage to persons or property arising from or related to any use of any material contained in JNeurosci.