Stanford Advisors


All Publications


  • Synaptic correlates of associative fear memory in the lateral amygdala NEURON Il Choi, D., Kim, J., Lee, H., Kim, J., Sung, Y., Choi, J., Venkat, S., Park, P., Jung, H., Kaang, B. 2021; 109 (17): 2717-+

    Abstract

    Successful adaptation to the environment requires an accurate response to external threats by recalling specific memories. Memory formation and recall require engram cell activity and synaptic strengthening among activated neuronal ensembles. However, elucidation of the underlying neural substrates of associative fear memory has remained limited without a direct interrogation of extinction-induced changes of specific synapses that encode a specific auditory fear memory. Using dual-eGRASP (enhanced green fluorescent protein reconstitution across synaptic partners), we found that synapses among activated neuronal ensembles or activated synaptic ensembles showed a significantly larger spine morphology at auditory cortex (AC)-to-lateral amygdala (LA) projections after auditory fear conditioning in mice. Fear extinction reversed these enhanced synaptic ensemble spines, whereas re-conditioning with the same tone and shock restored the spine size of the synaptic ensemble. We suggest that synaptic ensembles encode and represent different fear memory states.

    View details for DOI 10.1016/j.neuron.2021.07.003

    View details for Web of Science ID 000693584400009

    View details for PubMedID 34363751

  • Interregional synaptic maps among engram cells underlie memory formation SCIENCE Choi, J., Sim, S., Kim, J., Choi, D., Oh, J., Ye, S., Lee, J., Kim, T., Ko, H., Lim, C., Kaang, B. 2018; 360 (6387): 430–35

    Abstract

    Memory resides in engram cells distributed across the brain. However, the site-specific substrate within these engram cells remains theoretical, even though it is generally accepted that synaptic plasticity encodes memories. We developed the dual-eGRASP (green fluorescent protein reconstitution across synaptic partners) technique to examine synapses between engram cells to identify the specific neuronal site for memory storage. We found an increased number and size of spines on CA1 engram cells receiving input from CA3 engram cells. In contextual fear conditioning, this enhanced connectivity between engram cells encoded memory strength. CA3 engram to CA1 engram projections strongly occluded long-term potentiation. These results indicate that enhanced structural and functional connectivity between engram cells across two directly connected brain regions forms the synaptic correlate for memory formation.

    View details for DOI 10.1126/science.aas9204

    View details for Web of Science ID 000430949600041

    View details for PubMedID 29700265

  • Synaptic ensembles between raphe and D1R-containing accumbens shell neurons underlie postisolation sociability in males. Science advances Choi, J. E., Choi, D. I., Lee, J., Kim, J., Kim, M. J., Hong, I., Jung, H., Sung, Y., Kim, J. I., Kim, T., Yu, N. K., Lee, S. H., Choe, H. K., Koo, J. W., Kim, J. H., Kaang, B. K. 2022; 8 (41): eabo7527

    Abstract

    Social animals expend considerable energy to maintain social bonds throughout their life. Male and female mice show sexually dimorphic behaviors, yet the underlying neural mechanisms of sociability and their dysregulation during social disconnection remain unknown. Dopaminergic neurons in dorsal raphe nucleus (DRNTH) is known to contribute to a loneliness-like state and modulate sociability. We identified that activated subpopulations in DRNTH and nucleus accumbens shell (NAcsh) during 24 hours of social isolation underlie the increase in isolation-induced sociability in male but not in female mice. This effect was reversed by chemogenetically and optogenetically inhibiting the DRNTH-NAcsh circuit. Moreover, synaptic connectivity among the activated neuronal ensembles in this circuit was increased, primarily in D1 receptor-expressing neurons in NAcsh. The increase in synaptic density functionally correlated with elevated dopamine release into NAcsh. Overall, specific synaptic ensembles in DRNTH-NAcsh mediate sex differences in isolation-induced sociability, indicating that sex-dependent circuit dynamics underlie the expression of sexually dimorphic behaviors.

    View details for DOI 10.1126/sciadv.abo7527

    View details for PubMedID 36223467

  • Exogenous expression of an allatotropin-related peptide receptor increased the membrane excitability in Aplysia neurons MOLECULAR BRAIN Zhang, G., Guo, S., Yin, S., Yuan, W., Chen, P., Kim, J., Wang, H., Zhou, H., Susswein, A. J., Kaang, B., Jing, J. 2022; 15 (1): 42

    Abstract

    Neuropeptides act mostly on a class of G-protein coupled receptors, and play a fundamental role in the functions of neural circuits underlying behaviors. However, physiological functions of some neuropeptide receptors are poorly understood. Here, we used the molluscan model system Aplysia and microinjected the exogenous neuropeptide receptor apATRPR (Aplysia allatotropin-related peptide receptor) with an expression vector (pNEX3) into Aplysia neurons that did not express the receptor endogenously. Physiological experiments demonstrated that apATRPR could mediate the excitability increase induced by its ligand, apATRP (Aplysia allatotropin-related peptide), in the Aplysia neurons that now express the receptor. This study provides a definitive evidence for a physiological function of a neuropeptide receptor in molluscan animals.

    View details for DOI 10.1186/s13041-022-00929-4

    View details for Web of Science ID 000792612600002

    View details for PubMedID 35534865

    View details for PubMedCentralID PMC9082908

  • Loss of the neuronal genome organizer and transcription factorCTCFinduces neuronal death and reactive gliosis in the anterior cingulate cortex GENES BRAIN AND BEHAVIOR Kwak, J., Kim, S., Yu, N., Seo, H., Choi, J., Kim, J., Choi, D., Kim, M., Kwak, C., Lee, K., Kaang, B. 2021; 20 (2): e12701

    Abstract

    CCCTC-binding factor (CTCF) is a genome organizer that regulates gene expression through transcription and chromatin structure regulation. CTCF also plays an important role during the developmental and adult stages. Cell-specific CTCF deletion studies have shown that a reduction in CTCF expression leads to the development of distinct clinical features and cognitive disorders. Therefore, we knocked out Ctcf (CTCF cKO) in the excitatory neurons of the forebrain in a Camk2a-Cre mouse strain to examine the role of CTCF in cell death and gliosis in the cortex. CTCF cKO mice were viable, but they demonstrated an age-dependent increase in reactive gliosis of astrocytes and microglia in the anterior cingulate cortex (ACC) from 16 weeks of age prior to neuronal loss observed at over 20 weeks of age. Consistent with these data, qRT-PCR analysis of the CTCF cKO ACC revealed changes in the expression of inflammation-related genes (Hspa1a, Prokr2 and Itga8) linked to gliosis and neuronal death. Our results suggest that prolonged Ctcf gene deficiency in excitatory neurons results in neuronal cell death and gliosis, possibly through functional changes in inflammation-related genes.

    View details for DOI 10.1111/gbb.12701

    View details for Web of Science ID 000573380700001

    View details for PubMedID 32909350

  • Neur1andNeur2are required for hippocampus-dependent spatial memory and synaptic plasticity HIPPOCAMPUS Lee, J., Yoon, K., Park, P., Lee, C., Kim, M., Han, D., Kim, J., Kim, S., Lee, H., Lee, Y., Jang, E., Ko, H., Kong, Y., Kaang, B. 2020; 30 (11): 1158–66

    Abstract

    Neur1 and Neur2, mouse homologs of the Drosophila neur gene, consist of two neuralized homology repeat domains and a RING domain. Both Neur1 and Neur2 are expressed in the whole adult brain and encode E3 ubiquitin ligases, which play a crucial role in the Notch signaling pathways. A previous study reported that overexpression of Neur1 enhances hippocampus-dependent memory, whereas the role of Neur2 remains largely unknown. Here, we aimed to elucidate the respective roles of Neur1 and Neur2 in hippocampus-dependent memory using three lines of genetically modified mice: Neur1 knock-out, Neur2 knock-out, and Neur1 and Neur2 double knock-out (D-KO). Our results showed that spatial memory was impaired when both Neur1 and Neur2 were deleted, but not in the individual knock-out of either Neur1 or Neur2. In addition, basal synaptic properties estimated by input-output relationships and paired-pulse facilitation did not change, but a form of long-term potentiation that requires protein synthesis was specifically impaired in the D-KO mice. These results collectively suggest that Neur1 and Neur2 are crucially involved in hippocampus-dependent spatial memory and synaptic plasticity.

    View details for DOI 10.1002/hipo.23247

    View details for Web of Science ID 000546419000001

    View details for PubMedID 32644222

  • Overexpression of activated CaMKII in the CA1 hippocampus impairs context discrimination, but not contextual conditioning MOLECULAR BRAIN Ye, S., Kim, J., Kim, J., Kaang, B. 2019; 12: 32

    Abstract

    Calcium/Calmodulin-dependent protein kinase II (CaMKII) plays a key role in the molecular mechanism of memory formation. CaMKII is known to be activated specifically in the activated spines during memory formation. However, it is unclear whether the specific activation of CaMKII is necessary for encoding information. Here, we overexpressed active form of CaMKII (CaMKII*) in the hippocampal CA1 region to activate CaMKII nonspecifically. Moreover, we examined context-discrimination performance of mice. We found that the mice with overexpression of CaMKII* showed impaired context-discrimination ability, while the contextual fear conditioning remained intact. These results indicate that spatial specificity of CaMKII activation is necessary for context discrimination.

    View details for DOI 10.1186/s13041-019-0454-3

    View details for Web of Science ID 000463749900001

    View details for PubMedID 30953515

    View details for PubMedCentralID PMC6449978

  • In memoriam: John Lisman - commentaries on CaMKII as a memory molecule MOLECULAR BRAIN Bear, M. F., Cooke, S. F., Giese, K., Kaang, B., Kennedy, M. B., Kim, J., Morris, R. M., Park, P. 2018; 11: 76

    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.

    View details for DOI 10.1186/s13041-018-0419-y

    View details for Web of Science ID 000454531000002

    View details for PubMedID 30593282

    View details for PubMedCentralID PMC6309094

  • Remote Memory and Cortical Synaptic Plasticity Require Neuronal CCCTC-Binding Factor (CTCF) JOURNAL OF NEUROSCIENCE Kim, s., Yu, N., Shim, K., Kim, J., Kim, H., Han, D., Choi, J., Lee, S., Choi, D., Kim, M., Lee, D., Lee, K., Galjart, N., Lee, Y., Lee, J., Kaang, B. 2018; 38 (22): 5042–52

    Abstract

    The molecular mechanism of long-term memory has been extensively studied in the context of the hippocampus-dependent recent memory examined within several days. However, months-old remote memory maintained in the cortex for long-term has not been investigated much at the molecular level yet. Various epigenetic mechanisms are known to be important for long-term memory, but how the 3D chromatin architecture and its regulator molecules contribute to neuronal plasticity and systems consolidation is still largely unknown. CCCTC-binding factor (CTCF) is an 11-zinc finger protein well known for its role as a genome architecture molecule. Male conditional knock-out mice in which CTCF is lost in excitatory neurons during adulthood showed normal recent memory in the contextual fear conditioning and spatial water maze tasks. However, they showed remarkable impairments in remote memory in both tasks. Underlying the remote memory-specific phenotypes, we observed that female CTCF conditional knock-out mice exhibit disrupted cortical LTP, but not hippocampal LTP. Similarly, we observed that CTCF deletion in inhibitory neurons caused partial impairment of remote memory. Through RNA sequencing, we observed that CTCF knockdown in cortical neuron culture caused altered expression of genes that are highly involved in cell adhesion, synaptic plasticity, and memory. These results suggest that remote memory storage in the cortex requires CTCF-mediated gene regulation in neurons, whereas recent memory formation in the hippocampus does not.SIGNIFICANCE STATEMENT CCCTC-binding factor (CTCF) is a well-known 3D genome architectural protein that regulates gene expression. Here, we use two different CTCF conditional knock-out mouse lines and reveal, for the first time, that CTCF is critically involved in the regulation of remote memory. We also show that CTCF is necessary for appropriate expression of genes, many of which we found to be involved in the learning- and memory-related processes. Our study provides behavioral and physiological evidence for the involvement of CTCF-mediated gene regulation in the remote long-term memory and elucidates our understanding of systems consolidation mechanisms.

    View details for DOI 10.1523/JNEUROSCI.2738-17.2018

    View details for Web of Science ID 000435410700002

    View details for PubMedID 29712785

    View details for PubMedCentralID PMC6705941

  • Rapid Turnover of Cortical NCAM1 Regulates Synaptic Reorganization after Peripheral Nerve Injury CELL REPORTS Ko, H., Choi, J., Park, D., Kang, S., Lim, C., Sim, S., Shim, J., Kim, J., Kim, S., Choi, T., Ye, S., Lee, J., Park, P., Kim, S., Do, J., Park, J., Islam, M., Kim, H., Turck, C. W., Collingridge, G. L., Zhuo, M., Kaang, B. 2018; 22 (3): 748–59

    Abstract

    Peripheral nerve injury can induce pathological conditions that lead to persistent sensitized nociception. Although there is evidence that plastic changes in the cortex contribute to this process, the underlying molecular mechanisms are unclear. Here, we find that activation of the anterior cingulate cortex (ACC) induced by peripheral nerve injury increases the turnover of specific synaptic proteins in a persistent manner. We demonstrate that neural cell adhesion molecule 1 (NCAM1) is one of the molecules involved and show that it mediates spine reorganization and contributes to the behavioral sensitization. We show striking parallels in the underlying mechanism with the maintenance of NMDA-receptor- and protein-synthesis-dependent long-term potentiation (LTP) in the ACC. Our results, therefore, demonstrate a synaptic mechanism for cortical reorganization and suggest potential avenues for neuropathic pain treatment.

    View details for DOI 10.1016/j.celrep.2017.12.059

    View details for Web of Science ID 000423449400015

    View details for PubMedID 29346771

  • Strengthened connections between engrams encode specific memories BMB REPORTS Kim, J., Choi, D., Kaang, B. 2018; 51 (8): 369–70

    Abstract

    In previous studies, memory storage was localized to engram cells distributed across the brain. While these studies have provided an individual cellular profile of engram cells, their synaptic connectivity, or whether they follow Hebbian mechanisms, remains uncertain. Therefore, our recent study investigated whether synapses between engram cells exhibit selectively enhanced structural and functional properties following memory formation. This was accomplished using a newly developed technique called "dual-eGRASP". We found that the number and size of spines on CA1 engram cells that receive inputs from CA3 engram cells were larger than at other synapses. We further observed that this enhanced connectivity correlated with induced memory strength. CA3 engram synapses exhibited increased release probability, while CA1 engram synapses produced enhanced postsynaptic responses. CA3 engram to CA1 engram projections showed strong occlusion of long-term potentiation. We demonstrated that the synaptic connectivity of CA3 to CA1 engram cells was strengthened following memory formation. Our results suggest that Hebbian plasticity occurs during memory formation among engram cells at the synapse level. [BMB Reports 2018; 51(8): 369-370].

    View details for DOI 10.5483/BMBRep.2018.51.8.176

    View details for Web of Science ID 000452141700001

    View details for PubMedID 30078390

    View details for PubMedCentralID PMC6130830

  • The role of nuclear PKM zeta in memory maintenance NEUROBIOLOGY OF LEARNING AND MEMORY Ko, H., Kim, J., Sim, S., Kim, T., Yoo, J., Choi, S., Baek, S., Yu, W., Yoon, J., Sacktor, T., Kaang, B. 2016; 135: 50–56

    Abstract

    Recently, protein kinase M ζ (PKMζ) has emerged as an important player for maintaining memory. It has been reported that PKMζ regulates the trafficking of GluA2 in postsynaptic membranes to maintain memory. However, there has been no study on PKMζ outside the synaptic region regarding memory maintenance. Here, we found that PKMζ is transported to the nucleus in a neural activity-dependent manner. Moreover, we found that PKMζ phosphorylates CREB-binding protein (CBP) at serine residues and that PKMζ inhibition reduces the acetylation of histone H2B and H3. Finally, we showed that the amnesic effect of PKMζ inhibition can be rescued by enhancing histone acetylation level. These results suggest the possibility that nuclear PKMζ has a crucial role in memory maintenance.

    View details for DOI 10.1016/j.nlm.2016.06.010

    View details for Web of Science ID 000385054600007

    View details for PubMedID 27321162

    View details for PubMedCentralID PMC5500255

  • Which Neurons Will Be the Engram - Activated Neurons and/or More Excitable Neurons? EXPERIMENTAL NEUROBIOLOGY Kim, J., Cho, H., Han, J., Kaang, B. 2016; 25 (2): 55–63

    Abstract

    During past decades, the formation and storage principle of memory have received much attention in the neuroscience field. Although some studies have attempted to demonstrate the nature of the engram, elucidating the memory engram allocation mechanism was not possible because of the limitations of existing methods, which cannot specifically modulate the candidate neuronal population. Recently, the development of new techniques, which offer ways to mark and control specific populations of neurons, may accelerate solving this issue. Here, we review the recent advances, which have provided substantial evidence showing that both candidates (neuronal population that is activated by learning, and that has increased CREB level/excitability at learning) satisfy the criteria of the engram, which are necessary and sufficient for memory expression.

    View details for DOI 10.5607/en.2016.25.2.55

    View details for Web of Science ID 000406858300001

    View details for PubMedID 27122991

    View details for PubMedCentralID PMC4844563

  • Multiple repressive mechanisms in the hippocampus during memory formation SCIENCE Cho, J., Yu, N., Choi, J., Sim, S., Kang, S., Kwak, C., Lee, S., Kim, J., Choi, D., Kim, V., Kaang, B. 2015; 350 (6256): 82–87

    Abstract

    Memory stabilization after learning requires translational and transcriptional regulations in the brain, yet the temporal molecular changes that occur after learning have not been explored at the genomic scale. We used ribosome profiling and RNA sequencing to quantify the translational status and transcript levels in the mouse hippocampus after contextual fear conditioning. We revealed three types of repressive regulations: translational suppression of ribosomal protein-coding genes in the hippocampus, learning-induced early translational repression of specific genes, and late persistent suppression of a subset of genes via inhibition of estrogen receptor 1 (ESR1/ERα) signaling. In behavioral analyses, overexpressing Nrsn1, one of the newly identified genes undergoing rapid translational repression, or activating ESR1 in the hippocampus impaired memory formation. Collectively, this study unveils the yet-unappreciated importance of gene repression mechanisms for memory formation.

    View details for DOI 10.1126/science.aac7368

    View details for Web of Science ID 000362098300050

    View details for PubMedID 26430118