• Aucun résultat trouvé

General discussion and perspectives

Part 2. A synaptic mechanism of memory allocation

3. General discussion and perspectives

In the present work, the use of rodents together with tools for the remote control of neuronal activity (chemogenetic and optogenetics) during specific behavioral paradigms, revealed mechanisms that critically contribute to cellular engram formation.

We defined a new role of inhibitory neurotransmission in the selection of cellular engrams during hippocampal-dependent memory formation. To address this question, we used two different approaches applied to the DG of the hippocampus:

1. We studied different neuronal types responsible for GABAergic transmission.

2. We investigated the role of synaptic contacts that sense inhibitory neurotransmission.

Altogether, how the present study contributes to the current state-of-the-art of engram theory?

We have seen in the introduction that the theory of engram seems to have found already substantial evidence that sustain it (Tonegawa et al., 2015; Josselyn et al., 2015;

Eichenbaum, 2016). However, we also anticipated that many questions remain unanswered and different aspects of the cellular engrams need to be established and experimentally demonstrated. Some very instructive works suggest that cells are selected in the cellular engram based on their level of excitability and network activity (Han et al., 2007; Han et al., 2009; Yiu et al., 2014). However, a precise mechanism explaining how memory is allocated among individual neurons has not yet been revealed. This was exactly the aim of the present study where we could reveal two mechanisms that perform such function. We first revealed a network mechanism, a dendritic lateral inhibition among GCs mediated by SST+ INs, that determine the size of the cellular engram and the stability of the associated memory. In this first part, our results suggest that GCs are in competition between each other to become part of the cellular engram where active cells trigger SST+

INs activity that in turn prevent neighboring GCs to become part of it. Second, we showed that GABAergic synapses affect the size of the cellular engram and memory stability. This second mechanism, which seems to act mainly post-synaptically, suggests that GABAergic synapses are critical determinants for the formation of the cellular engram and affect memory function.

Altogether, we revealed a common phenomenon: the alteration of the size of the cellular engram that affects memory stability that was assessed by manipulating two different components of the inhibitory system of the DG: INs and GABAergic synapses.

What concerns arise and what are the missing gaps regarding studies on cellular engrams and the present study?

One of the major concern in studies investigating cellular engrams is the use of IEGs as neuronal markers. IEGs are one of the most used tools to study this question and their use has been demonstrated to be technically relevant and functional. Nevertheless, we should address some concerns about them.

First, what type of neuronal activity drives IEGs expression? According to the literature, there are only a few studies showing what exact pattern of activity lead to IEGs expression such as c-fos or Arc/Arg3.1. For example, we know that c-fos expression highly correlates to natural (Dragunow and Faull, 1989; Rakhade et al., 2007) or optogenetic induced neuronal firing (Schoenenberger et al., 2009). However, these studies could not provide extensive and robust evidence for the kind of activity necessary to induce c-fos expression. In our work, we revealed that optogenetic-induced neuronal firing at 10 Hz could induce c-fos expression in around 80% of the ChR2 expressing cells independently of their type (Figure 19D and 22B) suggesting that neuronal firing at this frequency could reliably induce c-fos expression. We could however not exclude that optogenetic induced back propagating action potentials as well as optogenetic-induced dendritic and membrane depolarization could alone or together induce c-fos expression.

Second, are IEGs expressed in the same way in different neuronal types involved in memory? When IEGs are used to identify recently activated neurons that belong to the cellular engram it is necessary to understand that c-fos expression may differ in different type of cells. There is consensus that neuronal activity induces c-fos expression in most of glutamatergic cells in many brain regions (Kovacs, 2008). However, it is known that other type of cells, especially INs, may have different signaling and cellular cascades that may differ in the expression of c-fos (Cohen et al., 2016). Indeed, in the present work, detection of activity-dependent induction of c-fos in PV+ and SST+ INs resulted to be less clear compared to excitatory neurons. For this reason, we used a complementary tool described in the results, a synaptic activity reporter (SARE), that permitted us to confirm the detection of recently activated PV+ and SST+ INs (Figure S4).

Third, is the temporal resolution of IEGs adequate to label cellular engram? Many agree about the fact that these genes are rapidly transcribed and reflect very recent changes in neuronal activity (Guzowski et al., 2005). However, their expression time course profile range form minutes to hours (15 minutes to 1-2 hours or more). Accordingly, their temporal resolution do not faithfully reflect their real activity, but rather a trace of it. Other currently

available tools would be more appropriate to sense real-time activity such as Ca2+

indicators for example, which may tell us a very different story about the activity of cells belonging to cellular engrams.

The use of opsins and designed receptors to control neuronal activity

Many studies, as well as the present work, use opsins and other proteins to artificially stimulate cells or group of neurons belonging to the cellular engram. In our study, the stimulation protocol used with optogenetics was 10Hz. This stimulation protocol, at least for the hippocampus, is often based to what it is observed in freely moving animals where place cells frequently attain this firing frequency (Haggerty and Ji, 2015; Passecker et al., 2011). However, it is also true that place cells are group of cells that increase their firing when the animal is in a particular location of the environment. In our study and in all the above mentioned that use optogenetics, it must be pointed out that we artificially forced all the cells expressing the opsin (in our study around 13%) to fire synchronously and at a fixed firing rate of 10Hz. This type of stimulation try to mimic at best what it is observed in physiological conditions where group of place cells (around 9% of cells for the DG (GoodSmith et al., 2017), while between 30% to 70% for CA1, (Wilson and McNaughton, 1993; Gothard et al., 1996), synchronize and organize spatial information. It is however necessary to keep in mind that we are still far to represent faithfully what really happens to the network when the animal encode a new memory. Moreover, a recent study reported that place cells in addition to the well-known spatial information also code for continuous, tasks-relevant variables (Aronov et al., 2017). Although artificial stimulation allows the animal to recall the associated memory trace, more sophisticated ways to manipulate neuronal activity that best matches natural conditions are needed.

Future perspectives

The very first step needed in studies investigating cellular engrams are the refinement of the tools used to identify, label and manipulate these populations of cells. Moreover, with the advances in the technology that we are assisting nowadays, it is very likely that in the near future some of the questions raised in the precedent paragraphs could find their answers. According to engram definitions, a population of cells, to acquire engram properties must satisfy different criteria (Josselyn et al., 2015; Guan et al., 2016). Most of the studies as well as the present one have been able to reveal only one or two criteria.

However, to really demonstrate the theory of engram one need to prove that the neuronal population studied satisfy all the criteria at the same time.

Most of the studies on memory have focused their attention either on the “spatial domain” by looking at specific brain regions, or on the “temporal domain” by investigating one particular stage of memory (i.e. encoding, consolidation, storage or recall). Thus, it would be impossible to give an answer to the question: “where is memory located in the brain?” Memory is a complex phenomenon combining the spatial and temporal domains.

Each stage is intrinsically connected with each other and should be considered as a continuum rather than separate systems capable of independent functioning. Accordingly, memory does not occur in one specific brain area but rather requires the interaction of many different brain regions. Recent studies have shown that oscillatory activity can synchronize firing in very distal regions of the brain and controls specific behaviors (Karalis et al., 2016; Dejean et al., 2016). For these reasons, further research in the field should consider the spatial and temporal domains together. A very recent study precisely stared this investigation and for the first time revealed that different brain areas are simultaneously involved in episodic memory formation and that they reorganize the mnemonic trace over time (Kitamura et al., 2017).

Most of the studies, as well as the present work, have revealed that a group of cells that are active during a specific behavior become part of the cellular engram. But what is the contribution of each single cell to the cellular engram? What is the information bearing in each single cell? Moreover, what is the role of cells that are not active (i.e. cells that are silenced by the active ensemble and do not express IEGs)? In our study, we have seen that lateral inhibition excludes other cells to become part of the cellular engram, but nothing is known about what these cells do. Do they “passively” participate to the cellular engram? Is synaptic plasticity occurring as well in these cells? It would be of major relevance in future experiments to investigate the role of these “passive” cells and understand their contribution to the cellular engram.

Moreover, memory plays a pivotal role in different cognitive processes that include emotion, attention, consciousness, decision, etc., all eventually affecting our behavior.

Indeed, many neuronal circuits are involved in these cognitive processes and interact with memory function. Emerging evidence reveals that dysfunctions in these neural circuits may cause brain pathologies that interfere with correct memory function. For these reasons, future research on memory need to combine all these different aspects (i.e.

cognitive processes, distinct brain regions and brain pathologies). A first step could be the refinement of memory/cognitive tasks. Most of the studies, including the present, have been using a very old but robust model to study episodic memory, the fear conditioning paradigm. We should highlight that although this task allowed great discoveries in the field

of cellular engram, it is however a very limited approach. Indeed, episodic memories are not limited to the mere fear memory but rather include a vast diversity of memories which may be associated with other internal states or emotions. Future experiments should provide more precise and complementary tasks together with animal models of brain disorders to gain insight in the function of memory.

To conclude, memory is a complex phenomenon implicating multiple brain regions. For this reason, memory research requires a brain-wide approach that allows activity recordings, gene-expression profile, cell-type and activity dependent connectivity tracing over the entire brain. It would be of interest in the very near future to use a combination of in vivo calcium imaging for population activity recordings, whole-brain connectivity visualization using clearing-tissue techniques and neuronal ensemble-tagging transgenic approach, to strengthen our understanding in this field. Furthermore, inhibitory circuits are known to be a key element in regulating interconnected excitatory networks and synchronize different brain areas. Accordingly, the knowledge revealed by the present study about inhibitory circuits will be extremely helpful for this future approach to the study memory. I believe that this holistic approach integrating observations made at different levels will lead to decisive contributions in the study of the brain mechanisms of memory.

REFERENCES

Acsady, L., Kamondi, A., Sik, A., Freund, T., and Buzsaki, G. (1998). GABAergic cells are the major postsynaptic targets of mossy fibers in the rat hippocampus. J Neurosci 18, 3386-3403.

Aggleton, J.P., and Pearce, J.M. (2001). Neural systems underlying episodic memory:

insights from animal research. Philos Trans R Soc Lond B Biol Sci 356, 1467-1482.

Alexander, G.M., Rogan, S.C., Abbas, A.I., Armbruster, B.N., Pei, Y., Allen, J.A., Nonneman, R.J., Hartmann, J., Moy, S.S., Nicolelis, M.A., et al. (2009). Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron 63, 27-39.

Alger, B.E., and Nicoll, R.A. (1979). GABA-mediated biphasic inhibitory responses in hippocampus. Nature 281, 315-317.

Altman, J., and Das, G.D. (1965). Post-natal origin of microneurones in the rat brain.

Nature 207, 953-956.

Amaral, D.G., Dolorfo, C., and Alvarez-Royo, P. (1991). Organization of CA1 projections to the subiculum: a PHA-L analysis in the rat. Hippocampus 1, 415-435.

Amaral, D.G., and Kurz, J. (1985). An analysis of the origins of the cholinergic and noncholinergic septal projections to the hippocampal formation of the rat. J Comp Neurol 240, 37-59.

Amaral, D.G., Scharfman, H.E., and Lavenex, P. (2007). The dentate gyrus: fundamental neuroanatomical organization (dentate gyrus for dummies). Prog Brain Res 163, 3-22.

Andersen, P., Dingledine, R., Gjerstad, L., Langmoen, I.A., and Laursen, A.M. (1980). Two different responses of hippocampal pyramidal cells to application of gamma-amino butyric acid. J Physiol 305, 279-296.

Anderson, M.I., and Jeffery, K.J. (2003). Heterogeneous modulation of place cell firing by changes in context. J Neurosci 23, 8827-8835.

Aronov, D., Nevers, R., and Tank, D.W. (2017). Mapping of a non-spatial dimension by the hippocampal-entorhinal circuit. Nature 543, 719-722.

Baisden, R.H., Woodruff, M.L., and Hoover, D.B. (1984). Cholinergic and non-cholinergic septo-hippocampal projections: a double-label horseradish peroxidase-acetylcholinesterase study in the rabbit. Brain Res 290, 146-151.

Barth, A.L., Gerkin, R.C., and Dean, K.L. (2004). Alteration of neuronal firing properties after in vivo experience in a FosGFP transgenic mouse. J Neurosci 24, 6466-6475.

Bartolomei, F., Barbeau, E., Gavaret, M., Guye, M., McGonigal, A., Regis, J., and Chauvel, P. (2004). Cortical stimulation study of the role of rhinal cortex in deja vu and reminiscence of memories. Neurology 63, 858-864.

Ben-Ari, Y. (2002). Excitatory actions of gaba during development: the nature of the nurture. Nat Rev Neurosci 3, 728-739.

Berger, T.K., Silberberg, G., Perin, R., and Markram, H. (2010). Brief bursts self-inhibit and correlate the pyramidal network. PLoS Biol 8.

Bergmann, O., Spalding, K.L., and Frisen, J. (2015). Adult Neurogenesis in Humans. Cold Spring Harb Perspect Biol 7, a018994.

Berridge, M.J. (1998). Neuronal calcium signaling. Neuron 21, 13-26.

Bliss, T.V., and Lomo, T. (1973). Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit following stimulation of the perforant path. J Physiol 232, 331-356.

Boccara, C.N., Sargolini, F., Thoresen, V.H., Solstad, T., Witter, M.P., Moser, E.I., and Moser, M.B. (2010). Grid cells in pre- and parasubiculum. Nat Neurosci 13, 987-994.

Bontempi, B., Laurent-Demir, C., Destrade, C., and Jaffard, R. (1999). Time-dependent reorganization of brain circuitry underlying long-term memory storage. Nature 400, 671-675.

Bormann, J. (2000). The 'ABC' of GABA receptors. Trends Pharmacol Sci 21, 16-19.

Bourne, J.N., and Harris, K.M. (2008). Balancing structure and function at hippocampal dendritic spines. Annu Rev Neurosci 31, 47-67.

Bramham, C.R., Worley, P.F., Moore, M.J., and Guzowski, J.F. (2008). The immediate early gene arc/arg3.1: regulation, mechanisms, and function. J Neurosci 28, 11760-11767.

Cai, D.J., Aharoni, D., Shuman, T., Shobe, J., Biane, J., Song, W., Wei, B., Veshkini, M., La-Vu, M., Lou, J., et al. (2016). A shared neural ensemble links distinct contextual memories encoded close in time. Nature 534, 115-118.

Cajal, S.R. (1894). The Croonian Lecture: La Fine Structure des Centres Nerveux. Proc R Soc Lond 55, 444-468.

Cameron, H.A., and McKay, R.D. (2001). Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus. J Comp Neurol 435, 406-417.

Chalifoux, J.R., and Carter, A.G. (2010). GABAB receptors modulate NMDA receptor calcium signals in dendritic spines. Neuron 66, 101-113.

Chen, S.X., Kim, A.N., Peters, A.J., and Komiyama, T. (2015). Subtype-specific plasticity of inhibitory circuits in motor cortex during motor learning. Nat Neurosci 18, 1109-1115.

Chiu, C.Q., Lur, G., Morse, T.M., Carnevale, N.T., Ellis-Davies, G.C., and Higley, M.J.

(2013). Compartmentalization of GABAergic inhibition by dendritic spines. Science 340, 759-762.

Choi, G.B., Stettler, D.D., Kallman, B.R., Bhaskar, S.T., Fleischmann, A., and Axel, R.

(2011). Driving opposing behaviors with ensembles of piriform neurons. Cell 146, 1004-1015.

Cichon, J., and Gan, W.B. (2015). Branch-specific dendritic Ca(2+) spikes cause persistent synaptic plasticity. Nature 520, 180-185.

Cirelli, C., and Tononi, G. (2000). On the functional significance of c-fos induction during the sleep-waking cycle. Sleep 23, 453-469.

Claiborne, B.J., Amaral, D.G., and Cowan, W.M. (1990). Quantitative, three-dimensional analysis of granule cell dendrites in the rat dentate gyrus. J Comp Neurol 302, 206-219.

Cohen, S.M., Ma, H., Kuchibhotla, K.V., Watson, B.O., Buzsaki, G., Froemke, R.C., and Tsien, R.W. (2016). Excitation-Transcription Coupling in Parvalbumin-Positive Interneurons Employs a Novel CaM Kinase-Dependent Pathway Distinct from Excitatory Neurons. Neuron 90, 292-307.

Cole, A.J., Saffen, D.W., Baraban, J.M., and Worley, P.F. (1989). Rapid increase of an immediate early gene messenger RNA in hippocampal neurons by synaptic NMDA receptor activation. Nature 340, 474-476.

Cossart, R., Dinocourt, C., Hirsch, J.C., Merchan-Perez, A., De Felipe, J., Ben-Ari, Y., Esclapez, M., and Bernard, C. (2001). Dendritic but not somatic GABAergic inhibition is decreased in experimental epilepsy. Nat Neurosci 4, 52-62.

Countryman, R.A., Kaban, N.L., and Colombo, P.J. (2005). Hippocampal c-fos is necessary for long-term memory of a socially transmitted food preference. Neurobiol Learn Mem 84, 175-183.

Cowansage, K.K., Shuman, T., Dillingham, B.C., Chang, A., Golshani, P., and Mayford, M.

(2014). Direct reactivation of a coherent neocortical memory of context. Neuron 84, 432-441.

Dejean, C., Courtin, J., Karalis, N., Chaudun, F., Wurtz, H., Bienvenu, T.C., and Herry, C.

(2016). Prefrontal neuronal assemblies temporally control fear behaviour. Nature 535, 420-424.

Deng, W., Aimone, J.B., and Gage, F.H. (2010). New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat Rev Neurosci 11, 339-350.

Deng, W., Mayford, M., and Gage, F.H. (2013). Selection of distinct populations of dentate granule cells in response to inputs as a mechanism for pattern separation in mice.

Elife 2, e00312.

Denny, C.A., Kheirbek, M.A., Alba, E.L., Tanaka, K.F., Brachman, R.A., Laughman, K.B., Tomm, N.K., Turi, G.F., Losonczy, A., and Hen, R. (2014). Hippocampal memory traces are differentially modulated by experience, time, and adult neurogenesis.

Neuron 83, 189-201.

Diamantaki, M., Frey, M., Berens, P., Preston-Ferrer, P., and Burgalossi, A. (2016).

Sparse activity of identified dentate granule cells during spatial exploration. Elife 5.

Dolleman-Van der Weel, M.J., and Witter, M.P. (2000). Nucleus reuniens thalami innervates gamma aminobutyric acid positive cells in hippocampal field CA1 of the rat. Neurosci Lett 278, 145-148.

Donato, F., Rompani, S.B., and Caroni, P. (2013). Parvalbumin-expressing basket-cell network plasticity induced by experience regulates adult learning. Nature 504, 272-276.

Dragunow, M., and Faull, R. (1989). The Use of C-Fos as a Metabolic Marker in Neuronal Pathway Tracing. Journal of Neuroscience Methods 29, 261-265.

Eichenbaum, H. (2016). Still searching for the engram. Learn Behav 44, 209-222.

Ekstrom, A.D., Kahana, M.J., Caplan, J.B., Fields, T.A., Isham, E.A., Newman, E.L., and Fried, I. (2003). Cellular networks underlying human spatial navigation. Nature 425, 184-188.

Estabrooke, I.V., McCarthy, M.T., Ko, E., Chou, T.C., Chemelli, R.M., Yanagisawa, M., Saper, C.B., and Scammell, T.E. (2001). Fos expression in orexin neurons varies with behavioral state. J Neurosci 21, 1656-1662.

Flavell, S.W., and Greenberg, M.E. (2008). Signaling mechanisms linking neuronal activity to gene expression and plasticity of the nervous system. Annu Rev Neurosci 31, 563-590.

Flores, C.E., and Mendez, P. (2014). Shaping inhibition: activity dependent structural plasticity of GABAergic synapses. Front Cell Neurosci 8, 327.

Flores, C.E., Nikonenko, I., Mendez, P., Fritschy, J.M., Tyagarajan, S.K., and Muller, D.

(2015). Activity-dependent inhibitory synapse remodeling through gephyrin phosphorylation. Proc Natl Acad Sci U S A 112, E65-72.

Frankland, P.W., and Bontempi, B. (2005). The organization of recent and remote memories. Nat Rev Neurosci 6, 119-130.

Frankland, P.W., Bontempi, B., Talton, L.E., Kaczmarek, L., and Silva, A.J. (2004). The involvement of the anterior cingulate cortex in remote contextual fear memory.

Science 304, 881-883.

Frankland, P.W., Ding, H.K., Takahashi, E., Suzuki, A., Kida, S., and Silva, A.J. (2006).

Stability of recent and remote contextual fear memory. Learn Mem 13, 451-457.

Frankland, P.W., Kohler, S., and Josselyn, S.A. (2013). Hippocampal neurogenesis and forgetting. Trends Neurosci 36, 497-503.

Freund, T.F., and Buzsaki, G. (1996). Interneurons of the hippocampus. Hippocampus 6, 347-470.

Freund, T.F., and Katona, I. (2007). Perisomatic inhibition. Neuron 56, 33-42.

Fritschy, J.M., Harvey, R.J., and Schwarz, G. (2008). Gephyrin: where do we stand, where do we go? Trends Neurosci 31, 257-264.

Funahashi, M., and Stewart, M. (1997). Presubicular and parasubicular cortical neurons of the rat: functional separation of deep and superficial neurons in vitro. J Physiol 501 ( Pt 2), 387-403.

Fyhn, M., Molden, S., Witter, M.P., Moser, E.I., and Moser, M.B. (2004). Spatial representation in the entorhinal cortex. Science 305, 1258-1264.

Gaiarsa, J.L., Caillard, O., and Ben-Ari, Y. (2002). Long-term plasticity at GABAergic and glycinergic synapses: mechanisms and functional significance. Trends Neurosci 25, 564-570.

Garner, A.R., Rowland, D.C., Hwang, S.Y., Baumgaertel, K., Roth, B.L., Kentros, C., and Mayford, M. (2012). Generation of a synthetic memory trace. Science 335, 1513-1516.

GoodSmith, D., Chen, X., Wang, C., Kim, S.H., Song, H., Burgalossi, A., Christian, K.M., and Knierim, J.J. (2017). Spatial Representations of Granule Cells and Mossy Cells of the Dentate Gyrus. Neuron 93, 677-690 e675.

Goold, C.P., and Nicoll, R.A. (2010). Single-cell optogenetic excitation drives homeostatic synaptic depression. Neuron 68, 512-528.

Gothard, K.M., Skaggs, W.E., and McNaughton, B.L. (1996). Dynamics of mismatch correction in the hippocampal ensemble code for space: interaction between path integration and environmental cues. J Neurosci 16, 8027-8040.