89 results on '"Picower Institute for Learning and Memory"'
Search Results
2. Three-dimensional chromatin organization in brain function and dysfunction
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Dileep, Vishnu, Tsai, Li-Huei, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Dileep, Vishnu, and Tsai, Li-Huei
- Abstract
The three-dimensional (3D) organization of chromatin within the nucleus is now recognized as a bona fide epigenetic property influencing genome function, replication, and maintenance. In the recent years, several studies have revealed how 3D chromatin organization is associated with brain function and its emerging role in disorders of the brain. 3D chromatin organization plays a crucial role in the development of different cell types of the nervous system and some neuronal cell types have adapted unique modifications to this organization that deviates from all other cell types. In post-mitotic neurons, dynamic changes in chromatin interactions in response to neuronal activity underlie learning and memory formation. Finally, new evidence directly links 3D chromatin organization to several disorders of the brain. These recent findings position 3D chromatin organization as a fundamental regulatory mechanism poised to reveal the etiology of brain function and dysfunctions., National Institute of Health (Grant 1-R01-NS102730-01)
- Published
- 2022
3. Spine Dynamics: Are They All the Same?
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Berry, Kalen Paul, Nedivi, Elly, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Berry, Kalen Paul, and Nedivi, Elly
- Abstract
© 2017 Elsevier Inc. Since Cajal's first drawings of Golgi stained neurons, generations of researchers have been fascinated by the small protrusions, termed spines, studding many neuronal dendrites. Most excitatory synapses in the mammalian CNS are located on dendritic spines, making spines convenient proxies for excitatory synaptic presence. When in vivo imaging revealed that dendritic spines are dynamic structures, their addition and elimination were interpreted as excitatory synapse gain and loss, respectively. Spine imaging has since become a popular assay for excitatory circuit remodeling. In this review, we re-evaluate the validity of using spine dynamics as a straightforward reflection of circuit rewiring. Recent studies tracking both spines and synaptic markers in vivo reveal that 20% of spines lack PSD-95 and are short lived. Although they account for most spine dynamics, their remodeling is unlikely to impact long-term network structure. We discuss distinct roles that spine dynamics can play in circuit remodeling depending on synaptic content. Dendritic spines are often viewed as proxies for glutamatergic synapses, and their addition or elimination as indicative of changes in circuit structure. In this review, Berry and Nedivi examine synaptic heterogeneity in the dendritic spine population and its implications for interpreting spine dynamics., NIH Pre-Doctoral Training Grant (T32GM007287), NIH grant (EY025437), NIH grant (EY011894)
- Published
- 2022
4. Imprinted Maternally Expressed microRNAs Antagonize Paternally Driven Gene Programs in Neurons
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Koch Institute for Integrative Cancer Research at MIT, Whipple, Amanda Joy, Breton-Provencher, Vincent, Jacobs, Hannah N., Patra, Chitta Ranjan, Sur, Mriganka, Sharp, Phillip A., Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Koch Institute for Integrative Cancer Research at MIT, Whipple, Amanda Joy, Breton-Provencher, Vincent, Jacobs, Hannah N., Patra, Chitta Ranjan, Sur, Mriganka, and Sharp, Phillip A.
- Abstract
Imprinted genes with parental-biased allelic expression are frequently co-regulated and enriched in common biological pathways. Here, we functionally characterize a large cluster of microRNAs (miRNAs) expressed from the maternally inherited allele (“maternally expressed”) to explore the molecular and cellular consequences of imprinted miRNA activity. Using an induced neuron (iN) culture system, we show that maternally expressed miRNAs from the miR-379/410 cluster direct the RNA-induced silencing complex (RISC) to transcriptional and developmental regulators, including paternally expressed transcripts like Plagl1. Maternal deletion of this imprinted miRNA cluster resulted in increased protein levels of several targets and upregulation of a broader transcriptional program regulating synaptic transmission and neuronal function. A subset of the transcriptional changes resulting from miR-379/410 deletion can be attributed to de-repression of Plagl1. These data suggest maternally expressed miRNAs antagonize paternally driven gene programs in neurons., National Institutes of Health (U.S.) (Grants P01-CA042063, R01-GM034277, R01-CA133404, R01-EY007023, R01-EY028219, R01-NS090473), National Cancer Institute (U.S.) (Grant P30-CA14051), National Institutes of Health (U.S.) (Postdoctoral fellowships NIH F32HD090833), National Institutes of Health (U.S.) (Grant FRQS 31677), Natural Sciences and Engineering Research Council Canada (Grant PDF-48724-2016)
- Published
- 2022
5. Selective Neuronal Vulnerability in Alzheimer’s Disease: A Network-Based Analysis
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Roussarie, Jean-Pierre, Yao, Vicky, Rodriguez-Rodriguez, Patricia, Oughtred, Rose, Rust, Jennifer, Plautz, Zakary, Kasturia, Shirin, Albornoz, Christian, Wang, Wei, Schmidt, Eric F, Dannenfelser, Ruth, Tadych, Alicja, Brichta, Lars, Barnea-Cramer, Alona, Heintz, Nathaniel, Hof, Patrick R, Heiman, Myriam, Dolinski, Kara, Flajolet, Marc, Troyanskaya, Olga G, Greengard, Paul, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Roussarie, Jean-Pierre, Yao, Vicky, Rodriguez-Rodriguez, Patricia, Oughtred, Rose, Rust, Jennifer, Plautz, Zakary, Kasturia, Shirin, Albornoz, Christian, Wang, Wei, Schmidt, Eric F, Dannenfelser, Ruth, Tadych, Alicja, Brichta, Lars, Barnea-Cramer, Alona, Heintz, Nathaniel, Hof, Patrick R, Heiman, Myriam, Dolinski, Kara, Flajolet, Marc, Troyanskaya, Olga G, and Greengard, Paul
- Abstract
© 2020 The Authors Neurons display different levels of vulnerability to Alzheimer's pathology. Roussarie et al. experimentally profile and computationally model several relevant neuron types. Using a mouse-human framework, they identify genes linking Aß, aging, and tau in vulnerable neurons. Finally, they show experimentally that PTB, a regulator of tau splicing, contributes to vulnerability.
- Published
- 2021
6. Extracellular Spike Waveform Dissociates Four Functionally Distinct Cell Classes in Primate Cortex
- Author
-
Picower Institute for Learning and Memory, Trainito, Caterina, von Nicolai, Constantin, Miller, Earl K, Siegel, Markus, Picower Institute for Learning and Memory, Trainito, Caterina, von Nicolai, Constantin, Miller, Earl K, and Siegel, Markus
- Abstract
© 2019 Elsevier Ltd Trainito et al. use a data-driven approach to robustly identify four cell classes from extracellular spike waveforms recorded in three cortical regions of macaque monkeys. The four cell classes are functionally distinct in terms of firing statistics, response dynamics, and information coding.
- Published
- 2021
7. Gamma Entrainment Binds Higher-Order Brain Regions and Offers Neuroprotection
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Media Laboratory, Massachusetts Institute of Technology. Department of Biological Engineering, Harvard University--MIT Division of Health Sciences and Technology, Adaikkan, Chinnakkaruppan, Middleton, Steven J, Marco, Asaf, Pao, Ping-Chieh, Mathys, Hansruedi, Kim, David Nam-Woo, Gao, Fan, Young, Jennie Z, Suk, Ho-Jun, Boyden, Edward S, McHugh, Thomas J, Tsai, Li-Huei, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Media Laboratory, Massachusetts Institute of Technology. Department of Biological Engineering, Harvard University--MIT Division of Health Sciences and Technology, Adaikkan, Chinnakkaruppan, Middleton, Steven J, Marco, Asaf, Pao, Ping-Chieh, Mathys, Hansruedi, Kim, David Nam-Woo, Gao, Fan, Young, Jennie Z, Suk, Ho-Jun, Boyden, Edward S, McHugh, Thomas J, and Tsai, Li-Huei
- Abstract
© 2019 Elsevier Inc. Chronic application of patterned visual stimulation in neurodegeneration mouse models to entrain gamma oscillations results in preservation of neuronal and synaptic density across multiple brain regions.
- Published
- 2021
8. Prefrontal oscillations modulate the propagation of neuronal activity required for working memory
- Author
-
Picower Institute for Learning and Memory, Sherfey, Jason, Ardid, Salva, Miller, Earl K, Hasselmo, Michael E, Kopell, Nancy J, Picower Institute for Learning and Memory, Sherfey, Jason, Ardid, Salva, Miller, Earl K, Hasselmo, Michael E, and Kopell, Nancy J
- Abstract
© 2020 The Author(s) Cognition involves using attended information, maintained in working memory (WM), to guide action. During a cognitive task, a correct response requires flexible, selective gating so that only the appropriate information flows from WM to downstream effectors that carry out the response. In this work, we used biophysically-detailed modeling to explore the hypothesis that network oscillations in prefrontal cortex (PFC), leveraging local inhibition, can independently gate responses to items in WM. The key role of local inhibition was to control the period between spike bursts in the outputs, and to produce an oscillatory response no matter whether the WM item was maintained in an asynchronous or oscillatory state. We found that the WM item that induced an oscillatory population response in the PFC output layer with the shortest period between spike bursts was most reliably propagated. The network resonant frequency (i.e., the input frequency that produces the largest response) of the output layer can be flexibly tuned by varying the excitability of deep layer principal cells. Our model suggests that experimentally-observed modulation of PFC beta-frequency (15–30 Hz) and gamma-frequency (30–80 Hz) oscillations could leverage network resonance and local inhibition to govern the flexible routing of signals in service to cognitive processes like gating outputs from working memory and the selection of rule-based actions. Importantly, we show for the first time that nonspecific changes in deep layer excitability can tune the output gate's resonant frequency, enabling the specific selection of signals encoded by populations in asynchronous or fast oscillatory states. More generally, this represents a dynamic mechanism by which adjusting network excitability can govern the propagation of asynchronous and oscillatory signals throughout neocortex.
- Published
- 2021
9. Microglia enable mature perineuronal nets disassembly upon anesthetic ketamine exposure or 60-Hz light entrainment in the healthy brain
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Venturino, Alessandro, Schulz, Rouven, De Jesús-Cortés, Héctor, Maes, Margaret E, Nagy, Bálint, Reilly-Andújar, Francis, Colombo, Gloria, Cubero, Ryan John A, Schoot Uiterkamp, Florianne E, Bear, Mark F, Siegert, Sandra, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Venturino, Alessandro, Schulz, Rouven, De Jesús-Cortés, Héctor, Maes, Margaret E, Nagy, Bálint, Reilly-Andújar, Francis, Colombo, Gloria, Cubero, Ryan John A, Schoot Uiterkamp, Florianne E, Bear, Mark F, and Siegert, Sandra
- Abstract
Perineuronal nets (PNNs), components of the extracellular matrix, preferentially coat parvalbumin-positive interneurons and constrain critical-period plasticity in the adult cerebral cortex. Current strategies to remove PNN are long-lasting, invasive, and trigger neuropsychiatric symptoms. Here, we apply repeated anesthetic ketamine as a method with minimal behavioral effect. We find that this paradigm strongly reduces PNN coating in the healthy adult brain and promotes juvenile-like plasticity. Microglia are critically involved in PNN loss because they engage with parvalbumin-positive neurons in their defined cortical layer. We identify external 60-Hz light-flickering entrainment to recapitulate microglia-mediated PNN removal. Importantly, 40-Hz frequency, which is known to remove amyloid plaques, does not induce PNN loss, suggesting microglia might functionally tune to distinct brain frequencies. Thus, our 60-Hz light-entrainment strategy provides an alternative form of PNN intervention in the healthy adult brain.
- Published
- 2021
10. Genome-wide In Vivo CNS Screening Identifies Genes that Modify CNS Neuronal Survival and mHTT Toxicity
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Wertz, Mary H, Mitchem, Mollie R, Pineda, S Sebastian, Hachigian, Lea J, Lee, Hyeseung, Lau, Vanessa, Powers, Alex, Kulicke, Ruth, Madan, Gurrein K, Colic, Medina, Therrien, Martine, Vernon, Amanda, Beja-Glasser, Victoria F, Hegde, Mudra, Gao, Fan, Kellis, Manolis, Hart, Traver, Doench, John G, Heiman, Myriam, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Wertz, Mary H, Mitchem, Mollie R, Pineda, S Sebastian, Hachigian, Lea J, Lee, Hyeseung, Lau, Vanessa, Powers, Alex, Kulicke, Ruth, Madan, Gurrein K, Colic, Medina, Therrien, Martine, Vernon, Amanda, Beja-Glasser, Victoria F, Hegde, Mudra, Gao, Fan, Kellis, Manolis, Hart, Traver, Doench, John G, and Heiman, Myriam
- Abstract
© 2020 Elsevier Inc. Unbiased in vivo genome-wide genetic screening is a powerful approach to elucidate new molecular mechanisms, but such screening has not been possible to perform in the mammalian central nervous system (CNS). Here, we report the results of the first genome-wide genetic screens in the CNS using both short hairpin RNA (shRNA) and CRISPR libraries. Our screens identify many classes of CNS neuronal essential genes and demonstrate that CNS neurons are particularly sensitive not only to perturbations to synaptic processes but also autophagy, proteostasis, mRNA processing, and mitochondrial function. These results reveal a molecular logic for the common implication of these pathways across multiple neurodegenerative diseases. To further identify disease-relevant genetic modifiers, we applied our screening approach to two mouse models of Huntington's disease (HD). Top mutant huntingtin toxicity modifier genes included several Nme genes and several genes involved in methylation-dependent chromatin silencing and dopamine signaling, results that reveal new HD therapeutic target pathways.
- Published
- 2021
11. Multi-sensory Gamma Stimulation Ameliorates Alzheimer’s-Associated Pathology and Improves Cognition
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Media Laboratory, Massachusetts Institute of Technology. Department of Biological Engineering, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Martorell, Anthony J, Paulson, Abigail L, Suk, Ho-Jun, Abdurrob, Fatema, Drummond, Gabrielle T, Guan, Webster, Young, Jennie Z, Kim, David Nam-Woo, Kritskiy, Oleg, Barker, Scarlett J, Mangena, Vamsi, Prince, Stephanie M, Brown, Emery N, Chung, Kwanghun, Boyden, Edward S, Singer, Annabelle C, Tsai, Li-Huei, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Media Laboratory, Massachusetts Institute of Technology. Department of Biological Engineering, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Chemical Engineering, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Martorell, Anthony J, Paulson, Abigail L, Suk, Ho-Jun, Abdurrob, Fatema, Drummond, Gabrielle T, Guan, Webster, Young, Jennie Z, Kim, David Nam-Woo, Kritskiy, Oleg, Barker, Scarlett J, Mangena, Vamsi, Prince, Stephanie M, Brown, Emery N, Chung, Kwanghun, Boyden, Edward S, Singer, Annabelle C, and Tsai, Li-Huei
- Abstract
© 2019 Elsevier Inc. We previously reported that inducing gamma oscillations with a non-invasive light flicker (gamma entrainment using sensory stimulus or GENUS) impacted pathology in the visual cortex of Alzheimer's disease mouse models. Here, we designed auditory tone stimulation that drove gamma frequency neural activity in auditory cortex (AC) and hippocampal CA1. Seven days of auditory GENUS improved spatial and recognition memory and reduced amyloid in AC and hippocampus of 5XFAD mice. Changes in activation responses were evident in microglia, astrocytes, and vasculature. Auditory GENUS also reduced phosphorylated tau in the P301S tauopathy model. Furthermore, combined auditory and visual GENUS, but not either alone, produced microglial-clustering responses, and decreased amyloid in medial prefrontal cortex. Whole brain analysis using SHIELD revealed widespread reduction of amyloid plaques throughout neocortex after multi-sensory GENUS. Thus, GENUS can be achieved through multiple sensory modalities with wide-ranging effects across multiple brain areas to improve cognitive function. Auditory stimulation combined with light-induced gamma oscillations in the hippocampus CA1 and auditory cortex regions of the brain reduces amyloid levels and improves memory in animal models of Alzheimer's disease.
- Published
- 2021
12. Cell Type-Specific Transcriptomics Reveals that Mutant Huntingtin Leads to Mitochondrial RNA Release and Neuronal Innate Immune Activation
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Lee, Hyeseung, Fenster, Robert J, Pineda, S Sebastian, Gibbs, Whitney S, Mohammadi, Shahin, Davila-Velderrain, Jose, Garcia, Francisco J, Therrien, Martine, Novis, Hailey S, Gao, Fan, Wilkinson, Hilary, Vogt, Thomas, Kellis, Manolis, LaVoie, Matthew J, Heiman, Myriam, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Lee, Hyeseung, Fenster, Robert J, Pineda, S Sebastian, Gibbs, Whitney S, Mohammadi, Shahin, Davila-Velderrain, Jose, Garcia, Francisco J, Therrien, Martine, Novis, Hailey S, Gao, Fan, Wilkinson, Hilary, Vogt, Thomas, Kellis, Manolis, LaVoie, Matthew J, and Heiman, Myriam
- Abstract
Lee, Fenster, et al. conduct striatal cell type-specific transcriptomics in mouse models of HD and human HD. Their findings reveal that mutant huntingtin leads to the release of mitochondrial RNAs and activation of innate immune signaling in neurons in a manner that correlates with striatal cell vulnerability in HD.
- Published
- 2021
13. GluN2A NMDA Receptor Enhancement Improves Brain Oscillations, Synchrony, and Cognitive Functions in Dravet Syndrome and Alzheimer’s Disease Models
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Hanson, Jesse E, Ma, Keran, Elstrott, Justin, Weber, Martin, Saillet, Sandrine, Khan, Abdullah S, Simms, Jeffrey, Liu, Benjamin, Kim, Thomas A, Yu, Gui-Qiu, Chen, Yelin, Wang, Tzu-Ming, Jiang, Zhiyu, Liederer, Bianca M, Deshmukh, Gauri, Solanoy, Hilda, Chan, Connie, Sellers, Benjamin D, Volgraf, Matthew, Schwarz, Jacob B, Hackos, David H, Weimer, Robby M, Sheng, Morgan, Gill, T Michael, Scearce-Levie, Kimberly, Palop, Jorge J, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Hanson, Jesse E, Ma, Keran, Elstrott, Justin, Weber, Martin, Saillet, Sandrine, Khan, Abdullah S, Simms, Jeffrey, Liu, Benjamin, Kim, Thomas A, Yu, Gui-Qiu, Chen, Yelin, Wang, Tzu-Ming, Jiang, Zhiyu, Liederer, Bianca M, Deshmukh, Gauri, Solanoy, Hilda, Chan, Connie, Sellers, Benjamin D, Volgraf, Matthew, Schwarz, Jacob B, Hackos, David H, Weimer, Robby M, Sheng, Morgan, Gill, T Michael, Scearce-Levie, Kimberly, and Palop, Jorge J
- Abstract
Hanson et al. examine the therapeutic effects of enhancing GluN2A-subunit-containing NMDAR function in Dravet syndrome and Alzheimer's disease mice. GNE-0723 treatment reduces aberrant low-frequency oscillations and epileptiform discharges and improves cognitive functions in both disease models. GluN2A NMDAR enhancers may benefit brain disorders with network hypersynchrony and cognitive impairments.
- Published
- 2021
14. Constructing a control-ready model of EEG signal during general anesthesia in humans
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Abel, John H, Badgeley, Marcus A, Baum, Taylor E, Chakravarty, Sourish, Purdon, Patrick L, Brown, Emery N, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Abel, John H, Badgeley, Marcus A, Baum, Taylor E, Chakravarty, Sourish, Purdon, Patrick L, and Brown, Emery N
- Abstract
Significant effort toward the automation of general anesthesia has been made in the past decade. One open challenge is in the development of control-ready patient models for closed-loop anesthesia delivery. Standard depth-of-anesthesia tracking does not readily capture inter-individual differences in response to anesthetics, especially those due to age, and does not aim to predict a relationship between a control input (infused anesthetic dose) and system state (commonly, a function of electroencephalography (EEG) signal). In this work, we developed a control-ready patient model for closed-loop propofol-induced anesthesia using data recorded during a clinical study of EEG during general anesthesia in ten healthy volunteers. We used principal component analysis to identify the low-dimensional state-space in which EEG signal evolves during anesthesia delivery. We parameterized the response of the EEG signal to changes in propofol target-site concentration using logistic models. We note that inter-individual differences in anesthetic sensitivity may be captured by varying a constant cofactor of the predicted effect-site concentration. We linked the EEG dose-response with the control input using a pharmacokinetic model. Finally, we present a simple nonlinear model predictive control in silico demonstration of how such a closed-loop system would work.
- Published
- 2021
15. Different Levels of Category Abstraction by Different Dynamics in Different Prefrontal Areas
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Wutz, Andreas, Loonis, Roman, Roy, Jefferson E, Donoghue, Jacob A, Miller, Earl K, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Wutz, Andreas, Loonis, Roman, Roy, Jefferson E, Donoghue, Jacob A, and Miller, Earl K
- Abstract
© 2018 Elsevier Inc. Categories can be grouped by shared sensory attributes (i.e., cats) or a more abstract rule (i.e., animals). We explored the neural basis of abstraction by recording from multi-electrode arrays in prefrontal cortex (PFC) while monkeys performed a dot-pattern categorization task. Category abstraction was varied by the degree of exemplar distortion from the prototype pattern. Different dynamics in different PFC regions processed different levels of category abstraction. Bottom-up dynamics (stimulus-locked gamma power and spiking) in the ventral PFC processed more low-level abstractions, whereas top-down dynamics (beta power and beta spike-LFP coherence) in the dorsal PFC processed more high-level abstractions. Our results suggest a two-stage, rhythm-based model for abstracting categories. Wutz et al. show that different levels of category abstraction engage different oscillatory dynamics in different prefrontal cortex (PFC) areas. This suggests a functional specialization within PFC for low-level, stimulus-based categories (e.g., cats) and high-level, rule-based categories (e.g., animals).
- Published
- 2021
16. A simulation-based comparative analysis of PID and LQG control for closed-loop anesthesia delivery
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Chakravarty, Sourish, Waite, Ayan S, Abel, John H, Brown, Emery N, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Chakravarty, Sourish, Waite, Ayan S, Abel, John H, and Brown, Emery N
- Abstract
Closed loop anesthesia delivery (CLAD) systems can help anesthesiologists efficiently achieve and maintain desired anesthetic depth over an extended period of time. A typical CLAD system would use an anesthetic marker, calculated from physiological signals, as real-time feedback to adjust anesthetic dosage towards achieving a desired set-point of the marker. Since control strategies for CLAD vary across the systems reported in recent literature, a comparative analysis of common control strategies can be useful. For a nonlinear plant model based on well-established models of compartmental pharmacokinetics and sigmoid-Emax pharmacodynamics, we numerically analyze the set-point tracking performance of three output-feedback linear control strategies: proportional-integral-derivative (PID) control, linear quadratic Gaussian (LQG) control, and an LQG with integral action (ILQG). Specifically, we numerically simulate multiple CLAD sessions for the scenario where the plant model parameters are unavailable for a patient and the controller is designed based on a nominal model and controller gains are held constant throughout a session. Based on the numerical analyses performed here, conditioned on our choice of model and controllers, we infer that in terms of accuracy and bias PID control performs better than ILQG which in turn performs better than LQG. In the case of noisy observations, ILQG can be tuned to provide a smoother infusion rate while achieving comparable steady state response with respect to PID. The numerical analysis framework and findings reported here can help CLAD developers in their choice of control strategies. This paper may also serve as a tutorial paper for teaching control theory for CLAD.
- Published
- 2021
17. Interhemispheric transfer of working memories
- Author
-
Picower Institute for Learning and Memory, Brincat, Scott Louis, Donoghue, Jacob Alexander, Mahnke, Meredith K., Kornblith, Simon, Lundqvist, Lars Mikael, Miller, Earl K, Picower Institute for Learning and Memory, Brincat, Scott Louis, Donoghue, Jacob Alexander, Mahnke, Meredith K., Kornblith, Simon, Lundqvist, Lars Mikael, and Miller, Earl K
- Abstract
Visual working memory (WM) storage is largely independent between the left and right visual hemifields/cerebral hemispheres, yet somehow WM feels seamless. We studied how WM is integrated across hemifields by recording neural activity bilaterally from lateral prefrontal cortex. An instructed saccade during the WM delay shifted the remembered location from one hemifield to the other. Before the shift, spike rates and oscillatory power showed clear signatures of memory laterality. After the shift, the lateralization inverted, consistent with transfer of the memory trace from one hemisphere to the other. Transferred traces initially used different neural ensembles from feedforward-induced ones, but they converged at the end of the delay. Around the time of transfer, synchrony between the two prefrontal hemispheres peaked in theta and beta frequencies, with a directionality consistent with memory trace transfer. This illustrates how dynamics between the two cortical hemispheres can stitch together WM traces across visual hemifields.Brincat et al. use bilateral recording to show working memories transferring between the right and left prefrontal cortex. Transferred memories engage different ensembles than feedforward-induced memory traces. Trace transfer is accompanied by directed interhemispheric theta/beta synchrony., NIMH (Grant R37MH087027), ONR (Grant MURI N00014-16-1-2832), NIGMS (Grant T32GM007753)
- Published
- 2021
18. Sensory processing and categorization in cortical and deep neural networks
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Pinotsis, Dimitrios, Miller, Earl K, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Pinotsis, Dimitrios, and Miller, Earl K
- Abstract
Many recent advances in artificial intelligence (AI) are rooted in visual neuroscience. However, ideas from more complicated paradigms like decision-making are less used. Although automated decision-making systems are ubiquitous (driverless cars, pilot support systems, medical diagnosis algorithms etc.), achieving human-level performance in decision making tasks is still a challenge. At the same time, these tasks that are hard for AI are easy for humans. Thus, understanding human brain dynamics during these decision-making tasks and modeling them using deep neural networks could improve AI performance. Here we modelled some of the complex neural interactions during a sensorimotor decision making task. We investigated how brain dynamics flexibly represented and distinguished between sensory processing and categorization in two sensory domains: motion direction and color. We used two different approaches for understanding neural representations. We compared brain responses to 1) the geometry of a sensory or category domain (domain selectivity) and 2) predictions from deep neural networks (computation selectivity). Both approaches gave us similar results. This confirmed the validity of our analyses. Using the first approach, we found that neural representations changed depending on context. We then trained deep recurrent neural networks to perform the same tasks as the animals. Using the second approach, we found that computations in different brain areas also changed flexibly depending on context. Color computations appeared to rely more on sensory processing, while motion computations more on abstract categories. Overall, our results shed light to the biological basis of categorization and differences in selectivity and computations in different brain areas. They also suggest a way for studying sensory and categorical representations in the brain: compare brain responses to both a behavioral model and a deep neural network and test if they give similar results., National Institute of Mental Health (U.S.) (Grant R37MH087027)
- Published
- 2021
19. Imprinted Maternally Expressed microRNAs Antagonize Paternally Driven Gene Programs in Neurons
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Koch Institute for Integrative Cancer Research at MIT, Whipple, Amanda Joy, Breton-Provencher, Vincent, Sur, Mriganka, Sharp, Phillip A., Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Koch Institute for Integrative Cancer Research at MIT, Whipple, Amanda Joy, Breton-Provencher, Vincent, Sur, Mriganka, and Sharp, Phillip A.
- Abstract
Imprinted genes with parental-biased allelic expression are frequently co-regulated and enriched in common biological pathways. Here, we functionally characterize a large cluster of microRNAs (miRNAs) expressed from the maternally inherited allele (“maternally expressed”) to explore the molecular and cellular consequences of imprinted miRNA activity. Using an induced neuron (iN) culture system, we show that maternally expressed miRNAs from the miR-379/410 cluster direct the RNA-induced silencing complex (RISC) to transcriptional and developmental regulators, including paternally expressed transcripts like Plagl1. Maternal deletion of this imprinted miRNA cluster resulted in increased protein levels of several targets and upregulation of a broader transcriptional program regulating synaptic transmission and neuronal function. A subset of the transcriptional changes resulting from miR-379/410 deletion can be attributed to de-repression of Plagl1. These data suggest maternally expressed miRNAs antagonize paternally driven gene programs in neurons., National Institutes of Health (U.S.) (Grants P01-CA042063, R01-GM034277, R01-CA133404, R01-EY007023, R01-EY028219, R01-NS090473), National Cancer Institute (U.S.) (Grant P30-CA14051), National Institutes of Health (U.S.) (Postdoctoral fellowships NIH F32HD090833), National Institutes of Health (U.S.) (Grant FRQS 31677), Natural Sciences and Engineering Research Council Canada (Grant PDF-48724-2016)
- Published
- 2021
20. Widespread Accumulation of Ribosome-Associated Isolated 3′ UTRs in Neuronal Cell Populations of the Aging Brain
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Biology, Sudmant, Peter, Lee, Hyeseung, Dominguez, Daniel, Heiman, Myriam, Burge, Christopher B., Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Biology, Sudmant, Peter, Lee, Hyeseung, Dominguez, Daniel, Heiman, Myriam, and Burge, Christopher B.
- Abstract
Particular brain regions and cell populations exhibit increased susceptibility to aging-related stresses. Here, we describe the age-specific and brain-region-specific accumulation of ribosome-associated 3′ UTR RNAs that lack the 5′ UTR and open reading frame. Our study reveals that this phenomenon impacts hundreds of genes in aged D1 spiny projection neurons of the mouse striatum and also occurs in the aging human brain. Isolated 3′ UTR accumulation is tightly correlated with mitochondrial gene expression and oxidative stress, with full-length mRNA expression that is reduced but not eliminated, and with production of short 3′ UTR-encoded peptides. Depletion of the oxidation-sensitive Fe-S cluster ribosome recycling factor ABCE1 induces the accumulation of 3′ UTRs, consistent with a model in which ribosome stalling and mRNA cleavage by No-Go decay yields isolated 3′ UTR RNAs protected by ribosomes. Isolated 3′ UTR accumulation is a hallmark of brain aging, likely reflecting regional differences in metabolism and oxidative stress. Particular brain regions and cell populations exhibit increased susceptibility to aging-related stresses. Sudmant et al. report that fragments of mRNAs accumulate in the aging brains of mice and humans. These species are associated with ribosomes and the production of small peptides and reflect regional differences in metabolism and oxidative stress., NIH (Grant HG002439)
- Published
- 2021
21. ASICs Mediate Food Responses in an Enteric Serotonergic Neuron that Controls Foraging Behaviors
- Author
-
Picower Institute for Learning and Memory, Rhoades, Jeffrey L., Nwabudike, Ijeoma, Yu, Stephanie K., McLachlan, Ian G., Madan, Gurrein K., Abebe, Eden, Powers, Joshua R., Flavell, Steven Willem, Picower Institute for Learning and Memory, Rhoades, Jeffrey L., Nwabudike, Ijeoma, Yu, Stephanie K., McLachlan, Ian G., Madan, Gurrein K., Abebe, Eden, Powers, Joshua R., and Flavell, Steven Willem
- Abstract
Animals must respond to the ingestion of food by generating adaptive behaviors, but the role of gut-brain signaling in behavioral regulation is poorly understood. Here, we identify conserved ion channels in an enteric serotonergic neuron that mediate its responses to food ingestion and decipher how these responses drive changes in foraging behavior. We show that the C. elegans serotonergic neuron NSM acts as an enteric sensory neuron that acutely detects food ingestion. We identify the novel and conserved acid-sensing ion channels (ASICs) DEL-7 and DEL-3 as NSM-enriched channels required for feeding-dependent NSM activity, which in turn drives slow locomotion while animals feed. Point mutations that alter the DEL-7 channel change NSM dynamics and associated behavioral dynamics of the organism. This study provides causal links between food ingestion, molecular and physiological properties of an enteric serotonergic neuron, and adaptive feeding behaviors, yielding a new view of how enteric neurons control behavior., University of Minnesota. Caenorhabditis Genetics Center (Grant P40 OD010440), National Institutes of Health (U.S.) (Grant R01NS104892), National Institutes of Health (U.S.) (Grant R01NS076558), National Science Foundation (U.S.) (Grant IOS 1353845)
- Published
- 2020
22. Functional implications of inhibitory synapse placement on signal processing in pyramidal neuron dendrites
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Department of Biology, Boivin, Josiah R., Nedivi, Elly, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Department of Biology, Boivin, Josiah R., and Nedivi, Elly
- Abstract
A rich literature describes inhibitory innervation of pyramidal neurons in terms of the distinct inhibitory cell types that target the soma, axon initial segment, or dendritic arbor. Less attention has been devoted to how localization of inhibition to specific parts of the pyramidal dendritic arbor influences dendritic signal detection and integration. The effect of inhibitory inputs can vary based on their placement on dendritic spines versus shaft, their distance from the soma, and the branch order of the dendrite they inhabit. Inhibitory synapses are also structurally dynamic, and the implications of these dynamics depend on their dendritic location. Here we consider the heterogeneous roles of inhibitory synapses as defined by their strategic placement on the pyramidal cell dendritic arbor., National Institutes of Health (Grants RO1 EY017656, RO1 EY025437, and F32 MH115441)
- Published
- 2020
23. Engram Cell Excitability State Determines the Efficacy of Memory Retrieval
- Author
-
RIKEN-MIT Center for Neural Circuit Genetics, Massachusetts Institute of Technology. Department of Biology, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Pignatelli di Spinazzola, Michele, Rivera, Tomás, Roy, Dheeraj, Lovett, Chanel W. L., Smith, Lillian, Muralidhar, Shruti, Tonegawa, Susumu, RIKEN-MIT Center for Neural Circuit Genetics, Massachusetts Institute of Technology. Department of Biology, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Pignatelli di Spinazzola, Michele, Rivera, Tomás, Roy, Dheeraj, Lovett, Chanel W. L., Smith, Lillian, Muralidhar, Shruti, and Tonegawa, Susumu
- Abstract
Animals need to optimize the efficacy of memory retrieval to adapt to environmental circumstances for survival. The recent development of memory engram labeling technology allows a precise investigation of the processes associated with the recall of a specific memory. Here, we show that engram cell excitability is transiently increased following memory reactivation. This short-term increase of engram excitability enhances the subsequent retrieval of specific memory content in response to cues and is manifest in the animal's ability to recognize contexts more precisely and more effectively. These results reveal a hitherto unknown transient enhancement of context recognition based on the plasticity of engram cell excitability. They also suggest that recall of a contextual memory is influenced by previous but recent activation of the same engram. The state of excitability of engram cells mediates differential behavioral outcomes upon memory retrieval and may be crucial for survival by promoting adaptive behavior.
- Published
- 2020
24. Npas4 Is a Critical Regulator of Learning-Induced Plasticity at Mossy Fiber-CA3 Synapses during Contextual Memory Formation
- Author
-
McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Harvard University--MIT Division of Health Sciences and Technology, Weng, Feng-Ju, Garcia, Rodrigo, Lutzu, Stefano, Alviña, Karina, Zhang, Yuxiang, Dushko, Margaret, Ku, Taeyun, Zemoura, Khaled, Rich, David, Garcia-Dominguez, Dario, Hung, Matthew, Yelhekar, Tushar D, Sørensen, Andreas Toft, Xu, Weifeng, Chung, Kwanghun, Castillo, Pablo E., Lin, Yingxi, McGovern Institute for Brain Research at MIT, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Department of Chemical Engineering, Harvard University--MIT Division of Health Sciences and Technology, Weng, Feng-Ju, Garcia, Rodrigo, Lutzu, Stefano, Alviña, Karina, Zhang, Yuxiang, Dushko, Margaret, Ku, Taeyun, Zemoura, Khaled, Rich, David, Garcia-Dominguez, Dario, Hung, Matthew, Yelhekar, Tushar D, Sørensen, Andreas Toft, Xu, Weifeng, Chung, Kwanghun, Castillo, Pablo E., and Lin, Yingxi
- Abstract
Synaptic connections between hippocampal mossy fibers (MFs) and CA3 pyramidal neurons are essential for contextual memory encoding, but the molecular mechanisms regulating MF-CA3 synapses during memory formation and the exact nature of this regulation are poorly understood. Here we report that the activity-dependent transcription factor Npas4 selectively regulates the structure and strength of MF-CA3 synapses by restricting the number of their functional synaptic contacts without affecting the other synaptic inputs onto CA3 pyramidal neurons. Using an activity-dependent reporter, we identified CA3 pyramidal cells that were activated by contextual learning and found that MF inputs on these cells were selectively strengthened. Deletion of Npas4 prevented both contextual memory formation and this learning-induced synaptic modification. We further show that Npas4 regulates MF-CA3 synapses by controlling the expression of the polo-like kinase Plk2. Thus, Npas4 is a critical regulator of experience-dependent, structural, and functional plasticity at MF-CA3 synapses during contextual memory formation. Weng et al. report that the transcription factor Npas4 selectively regulates the number of functional synaptic contacts between CA3 pyramidal neurons and mossy fibers, allowing for learning-induced modification of MF-CA3 synapses during contextual memory formation., NIH (Grants DA017392, NS090473, MH081935, MH091220, NS088421, and DC014701)
- Published
- 2020
25. A Library of Phosphoproteomic and Chromatin Signatures for Characterizing Cellular Responses to Drug Perturbations
- Author
-
Broad Institute of MIT and Harvard, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Litichevskiy, Lev, Peckner, Ryan, Abelin, Jennifer G., Asiedu, Jacob K., Creech, Amanda L., Davis, John F., Davison, Desiree, Dunning, Caitlin M., Egertson, Jarrett D., Egri, Shawn, Gould, Joshua, Ko, Tak, Johnson, Sarah A., Lahr, David L., Lam, Daniel, Liu, Zihan, MacLean, Brendan X., Lyons, Nicholas J., Lu, Xiaodong, Mungenast, Alison, Officer, Adam, Natoli, Ted E., Papanastasiou, Malvina, Patel, Jinal, Sharma, Vagisha, Toder, Courtney, Tubelli, Andrew A., Young, Jennie Zin-Ney, Carr, Steven A, Golub, Todd, Subramanian, Aravind, MacCoss, Michael J., Tsai, Li-Huei, Jaffe, Jacob D., Broad Institute of MIT and Harvard, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Litichevskiy, Lev, Peckner, Ryan, Abelin, Jennifer G., Asiedu, Jacob K., Creech, Amanda L., Davis, John F., Davison, Desiree, Dunning, Caitlin M., Egertson, Jarrett D., Egri, Shawn, Gould, Joshua, Ko, Tak, Johnson, Sarah A., Lahr, David L., Lam, Daniel, Liu, Zihan, MacLean, Brendan X., Lyons, Nicholas J., Lu, Xiaodong, Mungenast, Alison, Officer, Adam, Natoli, Ted E., Papanastasiou, Malvina, Patel, Jinal, Sharma, Vagisha, Toder, Courtney, Tubelli, Andrew A., Young, Jennie Zin-Ney, Carr, Steven A, Golub, Todd, Subramanian, Aravind, MacCoss, Michael J., Tsai, Li-Huei, and Jaffe, Jacob D.
- Abstract
Although the value of proteomics has been demonstrated, cost and scale are typically prohibitive, and gene expression profiling remains dominant for characterizing cellular responses to perturbations. However, high-throughput sentinel assays provide an opportunity for proteomics to contribute at a meaningful scale. We present a systematic library resource (90 drugs × 6 cell lines) of proteomic signatures that measure changes in the reduced-representation phosphoproteome (P100) and changes in epigenetic marks on histones (GCP). A majority of these drugs elicited reproducible signatures, but notable cell line- and assay-specific differences were observed. Using the “connectivity” framework, we compared signatures across cell types and integrated data across assays, including a transcriptional assay (L1000). Consistent connectivity among cell types revealed cellular responses that transcended lineage, and consistent connectivity among assays revealed unexpected associations between drugs. We further leveraged the resource against public data to formulate hypotheses for treatment of multiple myeloma and acute lymphocytic leukemia. This resource is publicly available at https://clue.io/proteomics. A large compendium of cellular responses to drugs as profiled through proteomic assays of phosphosignaling and histone modifications reveals cellular responses that transcend lineage, discovers unexpected associations between drugs, and recognizes therapeutic hypotheses for treatment of multiple myeloma and acute lymphocytic leukemia. Keywords: mass spectrometry; proteomics; drug discovery; signaling; epigenetics; mechanism of action; LINCS project; GCP; P100; L1000, NIH Common Fund's Library of Integrated Network-based Cellular Signatures (LINCS) program (Grant U54HG008097), NIH Common Fund's Library of Integrated Network-based Cellular Signatures (LINCS) program (Grant U54HG008699)
- Published
- 2020
26. Modular Assembly of Polysaccharide-Degrading Marine Microbial Communities
- Author
-
Massachusetts Institute of Technology. Department of Civil and Environmental Engineering, Massachusetts Institute of Technology. Computational and Systems Biology Program, Picower Institute for Learning and Memory, Enke, Tim N., Datta, Manoshi Sen, Schwartzman, Julia A., Cermak, Nathan, Cordero Sanchez, Otto X., Massachusetts Institute of Technology. Department of Civil and Environmental Engineering, Massachusetts Institute of Technology. Computational and Systems Biology Program, Picower Institute for Learning and Memory, Enke, Tim N., Datta, Manoshi Sen, Schwartzman, Julia A., Cermak, Nathan, and Cordero Sanchez, Otto X.
- Abstract
Understanding the principles that govern the assembly of microbial communities across earth's biomes is a major challenge in modern microbial ecology. This pursuit is complicated by the difficulties of mapping functional roles and interactions onto communities with immense taxonomic diversity and of identifying the scale at which microbes interact [1]. To address this challenge, here, we focused on the bacterial communities that colonize and degrade particulate organic matter in the ocean [2–4]. We show that the assembly of these communities can be simplified as a linear combination of functional modules. Using synthetic polysaccharide particles immersed in natural bacterioplankton assemblages [1, 5], we showed that successional particle colonization dynamics are driven by the interaction of two types of modules: a first type made of narrowly specialized primary degraders, whose dynamics are controlled by particle polysaccharide composition, and a second type containing substrate-independent taxa whose dynamics are controlled by interspecific interactions—in particular, cross-feeding via organic acids, amino acids, and other metabolic byproducts. We show that, as a consequence of this trophic structure, communities can assemble modularly—i.e., by a simple sum of substrate-specific primary degrader modules, one for each complex polysaccharide in the particle, connected to a single broad-niche range consumer module. Consistent with this model, a linear combination of the communities on single-polysaccharide particles accurately predicts community composition on mixed-polysaccharide particles. Our results suggest that the assembly of heterotrophic communities that degrade complex organic materials follows simple design principles that could be exploited to engineer heterotrophic microbiomes. Enke et al. show that particle-attached marine microbial communities assemble by recruiting functional groups of taxa in an additive manner. Specialist groups degrade specific poly, Simons Foundation. Simons Early Career Award (410104), Alfred P. Sloan Foundation. Fellowship (FG-20166236), National Science Foundation (U.S.) (Grant OCE-1658451), Simons Collaboration: Principles of Microbial Ecosystems (PriME) (Award 542395)
- Published
- 2020
27. A Meta-Analysis Suggests Different Neural Correlates for Implicit and Explicit Learning
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Loonis, Roman Florian, Brincat, Scott Louis, Antzoulatos, Evan G., Miller, Earl K, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Loonis, Roman Florian, Brincat, Scott Louis, Antzoulatos, Evan G., and Miller, Earl K
- Abstract
A meta-analysis of non-human primates performing three different tasks (Object-Match, Category-Match, and Category-Saccade associations) revealed signatures of explicit and implicit learning. Performance improved equally following correct and error trials in the Match (explicit) tasks, but it improved more after correct trials in the Saccade (implicit) task, a signature of explicit versus implicit learning. Likewise, error-related negativity, a marker for error processing, was greater in the Match (explicit) tasks. All tasks showed an increase in alpha/beta (10–30 Hz) synchrony after correct choices. However, only the implicit task showed an increase in theta (3–7 Hz) synchrony after correct choices that decreased with learning. In contrast, in the explicit tasks, alpha/beta synchrony increased with learning and decreased thereafter. Our results suggest that explicit versus implicit learning engages different neural mechanisms that rely on different patterns of oscillatory synchrony. Loonis et al. find that explicit and implicit learning use feedback about correct choices versus errors differently. Implicit learning relies more on theta synchrony (3–7 Hz) while explicit learning relies on alpha/beta synchrony (10–30 Hz). ©2017 Elsevier Inc., NIMH R37MH08702, NIMH R01MH06525, The Picower Institute Innovation Fund
- Published
- 2020
28. On memories, neural ensembles and mental flexibility
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Pinotsis, Dimitris A., Brincat, Scott Louis, Miller, Earl K, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Pinotsis, Dimitris A., Brincat, Scott Louis, and Miller, Earl K
- Abstract
Memories are assumed to be represented by groups of co-activated neurons, called neural ensembles. Describing ensembles is a challenge: complexity of the underlying micro-circuitry is immense. Current approaches use a piecemeal fashion, focusing on single neurons and employing local measures like pairwise correlations. We introduce an alternative approach that identifies ensembles and describes the effective connectivity between them in a holistic fashion. It also links the oscillatory frequencies observed in ensembles with the spatial scales at which activity is expressed. Using unsupervised learning, biophysical modeling and graph theory, we analyze multi-electrode LFPs from frontal cortex during a spatial delayed response task. We find distinct ensembles for different cues and more parsimonious connectivity for cues on the horizontal axis, which may explain the oblique effect in psychophysics. Our approach paves the way for biophysical models with learned parameters that can guide future Brain Computer Interface development. ©2017 Elsevier Inc., NIMH (R37MH087027), USAF grant (no. FA8655/13/1/3019)
- Published
- 2020
29. REST and Neural Gene Network Dysregulation in iPSC Models of Alzheimer’s Disease
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Meyer, Katharina, Feldman, Heather M., Lu, Tao, Drake, Derek, Lim, Elaine T., Ling, King-Hwa, Bishop, Nicholas A., Pan, Ying, Seo, Jinsoo, Lin, Yuan-Ta, Su, Susan C. (Susan Chih-Chieh), Church, George M., Tsai, Li-Huei, Yankner, Bruce A., Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Meyer, Katharina, Feldman, Heather M., Lu, Tao, Drake, Derek, Lim, Elaine T., Ling, King-Hwa, Bishop, Nicholas A., Pan, Ying, Seo, Jinsoo, Lin, Yuan-Ta, Su, Susan C. (Susan Chih-Chieh), Church, George M., Tsai, Li-Huei, and Yankner, Bruce A.
- Abstract
The molecular basis of the earliest neuronal changes that lead to Alzheimer’s disease (AD) is unclear. Here, we analyze neural cells derived from sporadic AD (SAD), APOE4 gene-edited and control induced pluripotent stem cells (iPSCs). We observe major differences in iPSC-derived neural progenitor (NP) cells and neurons in gene networks related to neuronal differentiation, neurogenesis, and synaptic transmission. The iPSC-derived neural cells from SAD patients exhibit accelerated neural differentiation and reduced progenitor cell renewal. Moreover, a similar phenotype appears in NP cells and cerebral organoids derived from APOE4 iPSCs. Impaired function of the transcriptional repressor REST is strongly implicated in the altered transcriptome and differentiation state. SAD and APOE4 expression result in reduced REST nuclear translocation and chromatin binding, and disruption of the nuclear lamina. Thus, dysregulation of neural gene networks may set in motion the pathologic cascade that leads to AD., National Institutes of Health (Grants R01AG046174, RF1AG048029)
- Published
- 2020
30. Corticoamygdala Transfer of Socially Derived Information Gates Observational Learning
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Research Laboratory of Electronics, Allsop, Stephen Azariah, Wichmann, Romy, Mills, Fergil, Burgos-Robles, Anthony Noel, Chang, Chia-Jung, Felix-Ortiz, Ada, Vienne, Alienor, Beyeler, Anna, Izadmehr, Ehsan, Glober, Gordon, Cum, Meghan I., Stergiadou, Johanna, Anandalingam, Kavitha, Farris, Kathryn, Namburi, Praneeth, Leppla, Christopher Albert, Weddington, Javier, Nieh, Horng-An Edward, Smith, Anne C., Ba, Demba E., Brown, Emery Neal, Tye, Kay M., Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Research Laboratory of Electronics, Allsop, Stephen Azariah, Wichmann, Romy, Mills, Fergil, Burgos-Robles, Anthony Noel, Chang, Chia-Jung, Felix-Ortiz, Ada, Vienne, Alienor, Beyeler, Anna, Izadmehr, Ehsan, Glober, Gordon, Cum, Meghan I., Stergiadou, Johanna, Anandalingam, Kavitha, Farris, Kathryn, Namburi, Praneeth, Leppla, Christopher Albert, Weddington, Javier, Nieh, Horng-An Edward, Smith, Anne C., Ba, Demba E., Brown, Emery Neal, and Tye, Kay M.
- Abstract
Observational learning is a powerful survival tool allowing individuals to learn about threat-predictive stimuli without directly experiencing the pairing of the predictive cue and punishment. This ability has been linked to the anterior cingulate cortex (ACC) and the basolateral amygdala (BLA). To investigate how information is encoded and transmitted through this circuit, we performed electrophysiological recordings in mice observing a demonstrator mouse undergo associative fear conditioning and found that BLA-projecting ACC (ACC→BLA) neurons preferentially encode socially derived aversive cue information. Inhibition of ACC→BLA alters real-time amygdala representation of the aversive cue during observational conditioning. Selective inhibition of the ACC→BLA projection impaired acquisition, but not expression, of observational fear conditioning. We show that information derived from observation about the aversive value of the cue is transmitted from the ACC to the BLA and that this routing of information is critically instructive for observational fear conditioning. Video Abstract: [Figure presented] For an individual to watch another's experience and learn from it, signals need to move from cortical neurons to the basolateral amygdala during detection and integration of the necessary social cues., NIMH (Grant R01-MH102441-01), NIA (Grant RF1-AG047661-01), NIDDK (Award DP2-DK-102256-01), NCCIH (Grant DP1-AT009925), NIH (Grants 1-R01-AG-050548-01, DP1-OD003646 and R01-GM104948)
- Published
- 2020
31. Unraveling the Paradox of Statins with Human Neurons: New Leads in Alzheimer’s Disease
- Author
-
Picower Institute for Learning and Memory, Blanchard, Joel W., Tsai, Li-Huei, Picower Institute for Learning and Memory, Blanchard, Joel W., and Tsai, Li-Huei
- Abstract
Conflicting clinical studies have reported that statins both reduce and accelerate cognitive impairments in Alzheimer’s disease. In this issue, Van der Kant et al. (2019) use iPSC-derived neurons to thoroughly dissect the link between cholesterol synthesis, phospho-Tau, and amyloid-β, revealing new therapeutic opportunities in Alzheimer’s disease and related dementias.
- Published
- 2019
32. A Developmental Switch in Microglial HDAC Function
- Author
-
Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Mathys, Hansruedi, Penney, Jay, Tsai, Li-Huei, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Mathys, Hansruedi, Penney, Jay, and Tsai, Li-Huei
- Abstract
The epigenetic mechanisms controlling microglia functions are largely unknown. In this issue of Immunity, Datta et al. (2018) uncover surprising and distinct effects of deleting the epigenetic regulators HDAC1 and HDAC2 during microglial development versus during the course of neurodegeneration.
- Published
- 2019
33. Deciphering Neural Codes of Memory during Sleep
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Chen, Zhe, Wilson, Matthew A., Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Chen, Zhe, and Wilson, Matthew A.
- Abstract
Memories of experiences are stored in the cerebral cortex. Sleep is critical for the consolidation of hippocampal memory of wake experiences into the neocortex. Understanding representations of neural codes of hippocampal–neocortical networks during sleep would reveal important circuit mechanisms in memory consolidation and provide novel insights into memory and dreams. Although sleep-associated ensemble spike activity has been investigated, identifying the content of memory in sleep remains challenging. Here we revisit important experimental findings on sleep-associated memory (i.e., neural activity patterns in sleep that reflect memory processing) and review computational approaches to the analysis of sleep-associated neural codes (SANCs). We focus on two analysis paradigms for sleep-associated memory and propose a new unsupervised learning framework (‘memory first, meaning later’) for unbiased assessment of SANCs. Keywords: sleep-associated memory; memory consolidation; memory replay; neural representation; population decoding; functional imaging, National Science Foundation (U.S.) (Award IIS-1307645), United States. Office of Naval Research (Grant N00014-10-1-0936), National Institutes of Health (U.S.) (Grant TR01-GM104948)
- Published
- 2019
34. In the loop: how chromatin topology links genome structure to function in mechanisms underlying learning and memory
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Watson, Lauren Ashley, Tsai, Li-Huei, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Watson, Lauren Ashley, and Tsai, Li-Huei
- Abstract
Different aspects of learning, memory, and cognition are regulated by epigenetic mechanisms such as covalent DNA modifications and histone post-translational modifications. More recently, the modulation of chromatin architecture and nuclear organization is emerging as a key factor in dynamic transcriptional regulation of the post-mitotic neuron. For instance, neuronal activity induces relocalization of gene loci to ‘transcription factories’, and specific enhancer–promoter looping contacts allow for precise transcriptional regulation. Moreover, neuronal activity-dependent DNA double-strand break formation in the promoter of immediate early genes appears to overcome topological constraints on transcription. Together, these findings point to a critical role for genome topology in integrating dynamic environmental signals to define precise spatiotemporal gene expression programs supporting cognitive processes.
- Published
- 2019
35. Basolateral to Central Amygdala Neural Circuits for Appetitive Behaviors
- Author
-
Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Kim, Joshua, Zhang, Xiangyu, Muralidhar, Shruti, LeBlanc, Sarah Anne, Tonegawa, Susumu, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Kim, Joshua, Zhang, Xiangyu, Muralidhar, Shruti, LeBlanc, Sarah Anne, and Tonegawa, Susumu
- Abstract
Basolateral amygdala (BLA) principal cells are capable of driving and antagonizing behaviors of opposing valence. BLA neurons project to the central amygdala (CeA), which also participates in negative and positive behaviors. However, the CeA has primarily been studied as the site for negative behaviors, and the causal role for CeA circuits underlying appetitive behaviors is poorly understood. Here, we identify several genetically distinct populations of CeA neurons that mediate appetitive behaviors and dissect the BLA-to-CeA circuit for appetitive behaviors. Protein phosphatase 1 regulatory subunit 1B⁺ BLA pyramidal neurons to dopamine receptor 1⁺ CeA neurons define a pathway for promoting appetitive behaviors, while R-spondin 2⁺ BLA pyramidal neurons to dopamine receptor 2⁺ CeA neurons define a pathway for suppressing appetitive behaviors. These data reveal genetically defined neural circuits in the amygdala that promote and suppress appetitive behaviors analogous to the direct and indirect pathways of the basal ganglia. Keywords: central amygdala; basolateral amygdala; direct and indirect pathways; appetitive; reward; fear; amygdala circuit; feeding; drinking; freezing, National Institutes of Health (U.S.) (Grant T32GM007287)
- Published
- 2018
36. Distinct Neural Circuits for the Formation and Retrieval of Episodic Memories
- Author
-
Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Roy, Dheeraj, Kitamura, Takashi, Okuyama, Teruhiro, Kitamura, Sachie Ogawa, Sun, Chen, Tonegawa, Susumu, Obata, Yuichi, Yoshiki, Atsushi, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Roy, Dheeraj, Kitamura, Takashi, Okuyama, Teruhiro, Kitamura, Sachie Ogawa, Sun, Chen, Tonegawa, Susumu, Obata, Yuichi, and Yoshiki, Atsushi
- Abstract
The formation and retrieval of a memory is thought to be accomplished by activation and reactivation, respectively, of the memory-holding cells (engram cells) by a common set of neural circuits, but this hypothesis has not been established. The medial temporal-lobe system is essential for the formation and retrieval of episodic memory for which individual hippocampal subfields and entorhinal cortex layers contribute by carrying out specific functions. One subfield whose function is poorly known is the subiculum. Here, we show that dorsal subiculum and the circuit, CA1 to dorsal subiculum to medial entorhinal cortex layer 5, play a crucial role selectively in the retrieval of episodic memories. Conversely, the direct CA1 to medial entorhinal cortex layer 5 circuit is essential specifically for memory formation. Our data suggest that the subiculum-containing detour loop is dedicated to meet the requirements associated with recall such as rapid memory updating and retrieval-driven instinctive fear responses., RIKEN Brain Science Institute, Howard Hughes Medical Institute, JPB Foundation
- Published
- 2018
37. Direct Medial Entorhinal Cortex Input to Hippocampal CA1 Is Crucial for Extended Quiet Awake Replay
- Author
-
Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Yamamoto, Jun, Tonegawa, Susumu, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Yamamoto, Jun, and Tonegawa, Susumu
- Abstract
Hippocampal replays have been demonstrated to play a crucial role in memory. Chains of ripples (ripple bursts) in CA1 have been reported to co-occur with long-range place cell sequence replays during the quiet awake state, but roles of neural inputs to CA1 in ripple bursts and replays are unknown. Here we show that ripple bursts in CA1 and medial entorhinal cortex (MEC) are temporally associated. An inhibition of MECIII input to CA1 during quiet awake reduced ripple bursts in CA1 and restricted the spatial coverage of replays to a shorter distance corresponding to single ripple events. The reduction did not occur with MECIII input inhibition during slow-wave sleep. Inhibition of CA3 activity suppressed ripples and replays in CA1 regardless of behavioral state. Thus, MECIII input to CA1 is crucial for ripple bursts and long-range replays specifically in quiet awake, whereas CA3 input is essential for both, regardless of behavioral state. Yamamoto and Tonegawa aim to determine the contribution of MECIII and CA3 inputs to hippocampal ripples and replays. They found differential roles of MECIII and CA3 inputs on CA1 ripples and replays during animal's different behavioral states., RIKEN Brain Science Institute, Howard Hughes Medical Institute, JPB Foundation
- Published
- 2018
38. Dissecting structure–activity-relationships of crebinostat: Brain penetrant HDAC inhibitors for neuroepigenetic regulation
- Author
-
Picower Institute for Learning and Memory, Tsai, Li-Huei, Ghosh, Balaram, Zhao, Wen-Ning, Reis, Surya A., Patnaik, Debasis, Fass, Daniel M., Mazitschek, Ralph, Haggarty, Stephen J., Picower Institute for Learning and Memory, Tsai, Li-Huei, Ghosh, Balaram, Zhao, Wen-Ning, Reis, Surya A., Patnaik, Debasis, Fass, Daniel M., Mazitschek, Ralph, and Haggarty, Stephen J.
- Abstract
Targeting chromatin-mediated epigenetic regulation has emerged as a potential avenue for developing novel therapeutics for a wide range of central nervous system disorders, including cognitive disorders and depression. Histone deacetylase (HDAC) inhibitors have been pursued as cognitive enhancers that impact the regulation of gene expression and other mechanisms integral to neuroplasticity. Through systematic modification of the structure of crebinostat, a previously discovered cognitive enhancer that affects genes critical to memory and enhances synaptogenesis, combined with biochemical and neuronal cell-based screening, we identified a novel hydroxamate-based HDAC inhibitor, here named neurinostat, with increased potency compared to crebinostat in inducing neuronal histone acetylation. In addition, neurinostat was found to have a pharmacokinetic profile in mouse brain modestly improved over that of crebinostat. This discovery of neurinostat and demonstration of its effects on neuronal HDACs adds to the available pharmacological toolkit for dissecting the molecular and cellular mechanisms of neuroepigenetic regulation in health and disease. Keywords: Cognitive enhancer; Nootropic; Histone deacetylases; Epigenetic; Chromatin; Acetylation; CREB; Neuroepigenetic
- Published
- 2018
39. Resolving CNS mRNA Heterogeneity: Examining mRNA Alternative Polyadenylation at a Cell-Type-Specific Level
- Author
-
Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Heiman, Myriam, Therrien, Martine, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Heiman, Myriam, and Therrien, Martine
- Abstract
Alternative polyadenylation often regulates mRNA isoform usage. In this issue of Neuron, Hwang et al. (2017) describe a powerful new cell-type-specific methodology, cTag-PAPERCLIP, which can be used to study alternative polyadenylation in the CNS. Keywords: alternative polyadenylation; cell type specificity; cTag-PAPERCLIP
- Published
- 2018
40. Imprinted Maternally Expressed microRNAs Antagonize Paternally Driven Gene Programs in Neurons
- Author
-
Udbhav K. Chitta, Phillip A. Sharp, Mriganka Sur, Vincent Breton-Provencher, Amanda J. Whipple, Hannah N. Jacobs, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
Transcription, Genetic ,Neurogenesis ,Biology ,Synaptic Transmission ,Cell Line ,Genomic Imprinting ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,microRNA ,Animals ,RNA-Induced Silencing Complex ,Gene silencing ,Allele ,Molecular Biology ,Gene ,Cells, Cultured ,Embryonic Stem Cells ,030304 developmental biology ,Neurons ,0303 health sciences ,Excitatory Postsynaptic Potentials ,Cell Biology ,Non-coding RNA ,3. Good health ,Cell biology ,MicroRNAs ,Argonaute Proteins ,Neuron differentiation ,Genomic imprinting ,Gene Deletion ,030217 neurology & neurosurgery - Abstract
Imprinted genes with parental-biased allelic expression are frequently co-regulated and enriched in common biological pathways. Here, we functionally characterize a large cluster of microRNAs (miRNAs) expressed from the maternally inherited allele (“maternally expressed”) to explore the molecular and cellular consequences of imprinted miRNA activity. Using an induced neuron (iN) culture system, we show that maternally expressed miRNAs from the miR-379/410 cluster direct the RNA-induced silencing complex (RISC) to transcriptional and developmental regulators, including paternally expressed transcripts like Plagl1. Maternal deletion of this imprinted miRNA cluster resulted in increased protein levels of several targets and upregulation of a broader transcriptional program regulating synaptic transmission and neuronal function. A subset of the transcriptional changes resulting from miR-379/410 deletion can be attributed to de-repression of Plagl1. These data suggest maternally expressed miRNAs antagonize paternally driven gene programs in neurons., National Institutes of Health (U.S.) (Grants P01-CA042063, R01-GM034277, R01-CA133404, R01-EY007023, R01-EY028219, R01-NS090473), National Cancer Institute (U.S.) (Grant P30-CA14051), National Institutes of Health (U.S.) (Postdoctoral fellowships NIH F32HD090833), National Institutes of Health (U.S.) (Grant FRQS 31677), Natural Sciences and Engineering Research Council Canada (Grant PDF-48724-2016)
- Published
- 2019
41. The Transcription Factor Sp3 Cooperates with HDAC2 to Regulate Synaptic Function and Plasticity in Neurons
- Author
-
Oleg Kritskiy, Fan Gao, Jun Wang, Li-Huei Tsai, Elizabeta Gjoneska, Satoko Yamakawa, Richard Rueda, Jay Penney, Hidekuni Yamakawa, Jemmie Cheng, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Yamakawa, Hidekuni, Cheng, Jemmie, Penney, Jay, Gao, Fan, Rueda IV, Richard, Wang, Jun, Yamakawa, Satoko, Kritskiy, Oleg, Gjoneska, Elizabeta, and Tsai, Li-Huei
- Subjects
Male ,0301 basic medicine ,Histone Deacetylase 2 ,Biology ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Histones ,Mice ,03 medical and health sciences ,Sp3 transcription factor ,Alzheimer Disease ,Memory ,Neuroplasticity ,medicine ,Animals ,Histone code ,lcsh:QH301-705.5 ,Neurons ,Gene knockdown ,synaptic plasticity ,Neuronal Plasticity ,Histone deacetylase 2 ,Neurodegeneration ,histone acetylation ,medicine.disease ,Histone Code ,Sp3 Transcription Factor ,030104 developmental biology ,lcsh:Biology (General) ,Synaptic plasticity ,Female ,Histone deacetylase ,Alzheimer’s disease ,Neuroscience ,epigenetic - Abstract
The histone deacetylase HDAC2, which negatively regulates synaptic gene expression and neuronal plasticity, is upregulated in Alzheimer’s disease (AD) patients and mouse models. Therapeutics targeting HDAC2 hold promise for ameliorating AD-related cognitive impairment; however, attempts to generate HDAC2-specific inhibitors have failed. Here, we take an integrative genomics approach to identify proteins that mediate HDAC2 recruitment to synaptic plasticity genes. Functional screening revealed that knockdown of the transcription factor Sp3 phenocopied HDAC2 knockdown and that Sp3 facilitated recruitment of HDAC2 to synaptic genes. Importantly, like HDAC2, Sp3 expression was elevated in AD patients and mouse models, where Sp3 knockdown ameliorated synaptic dysfunction. Furthermore, exogenous expression of an HDAC2 fragment containing the Sp3-binding domain restored synaptic plasticity and memory in a mouse model with severe neurodegeneration. Our findings indicate that targeting the HDAC2-Sp3 complex could enhance cognitive function without affecting HDAC2 function in other processes.
- Published
- 2017
42. In the loop: how chromatin topology links genome structure to function in mechanisms underlying learning and memory
- Author
-
L Ashley Watson, Li-Huei Tsai, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, and Picower Institute for Learning and Memory
- Subjects
0301 basic medicine ,Transcription factories ,Genome ,General Neuroscience ,Biology ,Topology ,Chromatin ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,030104 developmental biology ,Histone ,Spatiotemporal gene expression ,Memory ,Transcriptional regulation ,biology.protein ,Humans ,Learning ,Histone code ,Epigenetics ,Neuroscience ,Gene - Abstract
Different aspects of learning, memory, and cognition are regulated by epigenetic mechanisms such as covalent DNA modifications and histone post-translational modifications. More recently, the modulation of chromatin architecture and nuclear organization is emerging as a key factor in dynamic transcriptional regulation of the post-mitotic neuron. For instance, neuronal activity induces relocalization of gene loci to ‘transcription factories’, and specific enhancer–promoter looping contacts allow for precise transcriptional regulation. Moreover, neuronal activity-dependent DNA double-strand break formation in the promoter of immediate early genes appears to overcome topological constraints on transcription. Together, these findings point to a critical role for genome topology in integrating dynamic environmental signals to define precise spatiotemporal gene expression programs supporting cognitive processes.
- Published
- 2017
43. Sensory processing and categorization in cortical and deep neural networks
- Author
-
Dimitris A. Pinotsis, Earl K. Miller, Markus Siegel, Picower Institute for Learning and Memory, and Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences
- Subjects
Male ,Sensory processing ,Computer science ,Cognitive Neuroscience ,medicine.medical_treatment ,Decision Making ,Motion Perception ,BF ,Sensory system ,Context (language use) ,050105 experimental psychology ,Motion (physics) ,Article ,03 medical and health sciences ,0302 clinical medicine ,Deep Learning ,medicine ,Animals ,0501 psychology and cognitive sciences ,Categorical variable ,030304 developmental biology ,Cerebral Cortex ,0303 health sciences ,Artificial neural network ,Behavior, Animal ,business.industry ,05 social sciences ,Human brain ,Macaca mulatta ,3. Good health ,Task (computing) ,Recurrent neural network ,medicine.anatomical_structure ,Neurology ,Categorization ,RC0321 ,Female ,Artificial intelligence ,Nerve Net ,business ,030217 neurology & neurosurgery ,Color Perception ,Psychomotor Performance - Abstract
Many recent advances in artificial intelligence (AI) are rooted in visual neuroscience. However, ideas from more complicated paradigms like decision-making are less used. Although automated decision-making systems are ubiquitous (driverless cars, pilot support systems, medical diagnosis algorithms etc.), achieving human-level performance in decision making tasks is still a challenge. At the same time, these tasks that are hard for AI are easy for humans. Thus, understanding human brain dynamics during these decision-making tasks and modeling them using deep neural networks could improve AI performance. Here we modelled some of the complex neural interactions during a sensorimotor decision making task. We investigated how brain dynamics flexibly represented and distinguished between sensory processing and categorization in two sensory domains: motion direction and color. We used two different approaches for understanding neural representations. We compared brain responses to 1) the geometry of a sensory or category domain (domain selectivity) and 2) predictions from deep neural networks (computation selectivity). Both approaches gave us similar results. This confirmed the validity of our analyses. Using the first approach, we found that neural representations changed depending on context. We then trained deep recurrent neural networks to perform the same tasks as the animals. Using the second approach, we found that computations in different brain areas also changed flexibly depending on context. Color computations appeared to rely more on sensory processing, while motion computations more on abstract categories. Overall, our results shed light to the biological basis of categorization and differences in selectivity and computations in different brain areas. They also suggest a way for studying sensory and categorical representations in the brain: compare brain responses to both a behavioral model and a deep neural network and test if they give similar results., National Institute of Mental Health (U.S.) (Grant R37MH087027)
- Published
- 2019
44. Influence of midazolam premedication on intraoperative EEG signatures in elderly patients
- Author
-
Gregor Lichtner, Claudia Spies, Victoria Windmann, Devika Kishnan, Susanne Koch, Emery N. Brown, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, Massachusetts Institute of Technology. Institute for Data, Systems, and Society, Picower Institute for Learning and Memory, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, and Harvard University--MIT Division of Health Sciences and Technology
- Subjects
Male ,Intraoperative Neurophysiological Monitoring ,Midazolam ,Premedication ,Electroencephalography ,Anesthesia, General ,050105 experimental psychology ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,medicine ,Humans ,0501 psychology and cognitive sciences ,In patient ,Elective surgery ,Geriatric anesthesia ,Propofol ,Aged ,Cerebral Cortex ,medicine.diagnostic_test ,business.industry ,05 social sciences ,Age Factors ,Intraoperative EEG ,Sensory Systems ,Alpha Rhythm ,Neurology ,Anesthesia ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Anesthetics, Intravenous ,medicine.drug ,Adjuvants, Anesthesia - Abstract
Objective: To investigate the influence of midazolam premedication on the EEG-spectrum before and during general anesthesia in elderly patients. Methods: Patients aged ≥65 years, undergoing elective surgery were included in this prospective observational study. A continuous pre- and intraoperative frontal EEG was recorded in patients who received premedication with midazolam (Mid, n = 15) and patients who did not (noMid, n = 30). Absolute power within the delta (0.5–4 Hz), theta (4–8 Hz), alpha (8–12 Hz), and beta (12–25 Hz) frequency-bands was analyzed in EEG-sections before (pre-induction), and after induction of anesthesia with propofol (post-induction), as well as during general anesthesia with either propofol or volatile-anesthetics (intra-operative). Results: Pre-induction, α-power of Mid patients was lower compared with noMid-patients (α-power: Mid: −10.75 dB vs. noMid: −9.20 dB; p = 0.036). After induction of anesthesia Mid-patients displayed a stronger increase of frontal α-power resulting in higher absolute α-power at post-induction state, (α-power: Mid −3.56 dB vs. noMid: −6.69 dB; p = 0.004), which remained higher intraoperatively (α-power: Mid: −2.12 dB vs. noMid: −6.10 dB; p = 0.024). Conclusion: Midazolam premedication alters the intraoperative EEG-spectrum in elderly patients. Significance: This finding provides further evidence for the role of GABAergic activation in the induction of elevated, frontal α-power during general anesthesia. Keywords: Physiology (medical); Sensory Systems; Neurology; Clinical Neurology; Premedication; Benzodiazepines – midazolam; EEG; Geriatric anesthesia; Propofol anesthesia, Seventh Framework Programme (European Commission) (Grant HEALTH-F2-2014-60246)
- Published
- 2019
45. Interhemispheric transfer of working memories
- Author
-
Mikael Lundqvist, Simon Kornblith, Scott L. Brincat, Earl K. Miller, Jacob A. Donoghue, Meredith Mahnke, and Picower Institute for Learning and Memory
- Subjects
Male ,0301 basic medicine ,genetic structures ,Working memory ,General Neuroscience ,Prefrontal Cortex ,Cognition ,Engram ,Macaca mulatta ,Functional Laterality ,Lateralization of brain function ,03 medical and health sciences ,Memory, Short-Term ,030104 developmental biology ,0302 clinical medicine ,Laterality ,Saccade ,Visual Perception ,Animals ,Directionality ,Female ,Prefrontal cortex ,Psychology ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Visual working memory (WM) storage is largely independent between the left and right visual hemifields/cerebral hemispheres, yet somehow WM feels seamless. We studied how WM is integrated across hemifields by recording neural activity bilaterally from lateral prefrontal cortex. An instructed saccade during the WM delay shifted the remembered location from one hemifield to the other. Before the shift, spike rates and oscillatory power showed clear signatures of memory laterality. After the shift, the lateralization inverted, consistent with transfer of the memory trace from one hemisphere to the other. Transferred traces initially used different neural ensembles from feedforward-induced ones, but they converged at the end of the delay. Around the time of transfer, synchrony between the two prefrontal hemispheres peaked in theta and beta frequencies, with a directionality consistent with memory trace transfer. This illustrates how dynamics between the two cortical hemispheres can stitch together WM traces across visual hemifields.Brincat et al. use bilateral recording to show working memories transferring between the right and left prefrontal cortex. Transferred memories engage different ensembles than feedforward-induced memory traces. Trace transfer is accompanied by directed interhemispheric theta/beta synchrony., NIMH (Grant R37MH087027), ONR (Grant MURI N00014-16-1-2832), NIGMS (Grant T32GM007753)
- Published
- 2021
46. Bidirectional modulation of anxiety-related and social behaviors by amygdala projections to the medial prefrontal cortex
- Author
-
Neha D. Bhagat, Ada C. Felix-Ortiz, Anthony Burgos-Robles, Kay M. Tye, Christopher A. Leppla, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Felix-Ortiz, Ada Celis, Burgos-Robles, Anthony Noel, Bhagat, Neha D., Leppla, Christopher Albert, and Tye, Kay
- Subjects
Male ,0301 basic medicine ,Elevated plus maze ,Neuroscience(all) ,Prefrontal Cortex ,Anxiety ,Motor Activity ,Optogenetics ,infralimbic ,behavioral disciplines and activities ,Amygdala ,Article ,Open field ,stress ,03 medical and health sciences ,0302 clinical medicine ,anxiety disorders ,medicine ,Animals ,optogenetics ,Maze Learning ,Social Behavior ,Prefrontal cortex ,prelimbic ,General Neuroscience ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Anxiogenic ,fear ,Psychology ,Proto-Oncogene Proteins c-fos ,Neuroscience ,psychological phenomena and processes ,030217 neurology & neurosurgery ,Social behavior ,Basolateral amygdala - Abstract
The basolateral amygdala (BLA) and the medial prefrontal cortex (mPFC) modulate anxiety and social behaviors. It remains to be elucidated, however, whether direct projections from the BLA to the mPFC play a functional role in these behaviors. We used optogenetic approaches in behaving mice to either activate or inhibit BLA inputs to the mPFC during behavioral assays that assess anxiety-like behavior and social interaction. Channelrhodopsin-2 (ChR2)-mediated activation of BLA inputs to the mPFC produced anxiogenic effects in the elevated plus maze and open field test, whereas halorhodopsin (NpHR)-mediated inhibition produced anxiolytic effects. Furthermore, activation of the BLA-mPFC pathway reduced social interaction in the resident-intruder test, whereas inhibition facilitated social interaction. These results establish a causal relationship between activity in the BLA-mPFC pathway and the bidirectional modulation of anxiety-related and social behaviors., National Institutes of Health (U.S.) (National Research Service Award Institutional Research Training Grant 5T32GM007484-38), Brain & Behavior Research Foundation (Young Investigator Award), National Institute of Mental Health (U.S.) (Research Supplement to Promote Diversity in Health-Related Sciences), Integrative Neuronal Systems Fellowship, James R. Killian Fellowship, JPB Foundation, Whitehall Foundation, Klingenstein Foundation, Alfred P. Sloan Foundation, New York Stem Cell Foundation, National Institutes of Health (U.S.) (R01-MH102441-01)
- Published
- 2016
47. A Bayesian nonparametric approach for uncovering rat hippocampal population codes during spatial navigation
- Author
-
Matthew J. Johnson, Matthew A. Wilson, Zhe Chen, Scott W. Linderman, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, and Wilson, Matthew A
- Subjects
0301 basic medicine ,Hyperparameter ,Quantitative Biology::Neurons and Cognition ,business.industry ,Computer science ,General Neuroscience ,Markov chain Monte Carlo ,Bayesian inference ,Machine learning ,computer.software_genre ,Variable-order Bayesian network ,Statistics::Computation ,Bayesian statistics ,03 medical and health sciences ,symbols.namesake ,Bayes' theorem ,030104 developmental biology ,0302 clinical medicine ,symbols ,Artificial intelligence ,business ,Particle filter ,computer ,030217 neurology & neurosurgery ,Gibbs sampling - Abstract
Background: Rodent hippocampal population codes represent important spatial information about the environment during navigation. Computational methods have been developed to uncover the neural representation of spatial topology embedded in rodent hippocampal ensemble spike activity. New method: We extend our previous work and propose a novel Bayesian nonparametric approach to infer rat hippocampal population codes during spatial navigation. To tackle the model selection problem, we leverage a Bayesian nonparametric model. Specifically, we apply a hierarchical Dirichlet process-hidden Markov model (HDP-HMM) using two Bayesian inference methods, one based on Markov chain Monte Carlo (MCMC) and the other based on variational Bayes (VB). Results: The effectiveness of our Bayesian approaches is demonstrated on recordings from a freely behaving rat navigating in an open field environment. Comparison with existing methods: The HDP-HMM outperforms the finite-state HMM in both simulated and experimental data. For HPD-HMM, the MCMC-based inference with Hamiltonian Monte Carlo (HMC) hyperparameter sampling is flexible and efficient, and outperforms VB and MCMC approaches with hyperparameters set by empirical Bayes. Conclusion: The Bayesian nonparametric HDP-HMM method can efficiently perform model selection and identify model parameters, which can used for modeling latent-state neuronal population dynamics., National Institutes of Health (U.S.) (Grant R01-MH06197), National Institutes of Health (U.S.) (Grant TR01-GM10498), National Institutes of Health (U.S.) (Grant ONR-MURI N00014-10-1-0936)
- Published
- 2016
48. Simple, Scalable Proteomic Imaging for High-Dimensional Profiling of Intact Systems
- Author
-
Jeong Yoon Park, Kwanghun Chung, Justin Swaney, Sung-Yon Kim, Daniel Goodwin, Jae Hun Cho, Van J. Wedeen, Young Gyun Park, Matthew P. Frosch, H. Sebastian Seung, Margaret McCue, Austin Hubbert, Taeyun Ku, Sara Vassallo, Heejin Choi, Naveed A. Bakh, Evan Murray, Institute for Medical Engineering and Science, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Department of Chemical Engineering, Picower Institute for Learning and Memory, Murray, Evan, Cho, Jae Hun, Ku, Taeyun, Swaney, Justin Mark, Kim, Sung-Yon, Choi, Heejin, Park, Young-Gyun, Park, Jeong-Yoon, Hubbert, Austin W., McCue, Margaret Grace, Ling, Sara Lynn, Bakh, Naveed Ali, and Chung, Kwanghun
- Subjects
Male ,Proteomics ,Tissue architecture ,Software_GENERAL ,ComputingMethodologies_SIMULATIONANDMODELING ,ComputingMethodologies_IMAGEPROCESSINGANDCOMPUTERVISION ,High dimensional ,Computational biology ,Biology ,Bioinformatics ,Nerve Fibers, Myelinated ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Profiling (information science) ,ComputingMethodologies_COMPUTERGRAPHICS ,030304 developmental biology ,0303 health sciences ,Tissue Preservation ,Biochemistry, Genetics and Molecular Biology(all) ,Protein Expression Profiling ,Molecular Imaging ,3. Good health ,Mice, Inbred C57BL ,ComputingMethodologies_PATTERNRECOGNITION ,Reducing Agents ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Scalability ,Female ,Molecular imaging ,Algorithms ,030217 neurology & neurosurgery - Abstract
Combined measurement of diverse molecular and anatomical traits that span multiple levels remains a major challenge in biology. Here, we introduce a simple method that enables proteomic imaging for scalable, integrated, high-dimensional phenotyping of both animal tissues and human clinical samples. This method, termed SWITCH, uniformly secures tissue architecture, native biomolecules, and antigenicity across an entire system by synchronizing the tissue preservation reaction. The heat- and chemical-resistant nature of the resulting framework permits multiple rounds (>20) of relabeling. We have performed 22 rounds of labeling of a single tissue with precise co-registration of multiple datasets. Furthermore, SWITCH synchronizes labeling reactions to improve probe penetration depth and uniformity of staining. With SWITCH, we performed combinatorial protein expression profiling of the human cortex and also interrogated the geometric structure of the fiber pathways in mouse brains. Such integrated high-dimensional information may accelerate our understanding of biological systems at multiple levels., Simons Foundation. Postdoctoral Fellowship, Life Sciences Research Foundation, Burroughs Wellcome Fund (Career Award at the Scientific Interface), Searle Scholars Program, Michael J. Fox Foundation for Parkinson's Research, United States. Defense Advanced Research Projects Agency, National Institutes of Health (U.S.) (1-U01-NS090473-01)
- Published
- 2015
49. Memory engram storage and retrieval
- Author
-
Michele Pignatelli, Dheeraj S. Roy, Tomás J. Ryan, Susumu Tonegawa, Picower Institute for Learning and Memory, Tonegawa, Susumu, Pignatelli di Spinazzola, Michele, Roy, Dheeraj, and Ryan, Tomas John
- Subjects
Neurons ,Neuronal Plasticity ,Hardware_MEMORYSTRUCTURES ,Recall ,Long-term memory ,Neuroscience(all) ,General Neuroscience ,Amnesia ,Engram ,Spatial memory ,Optogenetics ,Memory ,Mental Recall ,Neuroplasticity ,Explicit memory ,medicine ,Animals ,Humans ,Amnesia, Retrograde ,Memory consolidation ,Nerve Net ,medicine.symptom ,Psychology ,Genes, Immediate-Early ,Neuroscience - Abstract
A great deal of experimental investment is directed towards questions regarding the mechanisms of memory storage. Such studies have traditionally been restricted to investigation of the anatomical structures, physiological processes, and molecular pathways necessary for the capacity of memory storage, and have avoided the question of how individual memories are stored in the brain. Memory engram technology allows the labeling and subsequent manipulation of components of specific memory engrams in particular brain regions, and it has been established that cell ensembles labeled by this method are both sufficient and necessary for memory recall. Recent research has employed this technology to probe fundamental questions of memory consolidation, differentiating between mechanisms of memory retrieval from the true neurobiology of memory storage.
- Published
- 2015
50. Widespread Accumulation of Ribosome-Associated Isolated 3′ UTRs in Neuronal Cell Populations of the Aging Brain
- Author
-
Myriam Heiman, Daniel Dominguez, Hyeseung Lee, Peter H. Sudmant, Christopher B. Burge, Picower Institute for Learning and Memory, and Massachusetts Institute of Technology. Department of Biology
- Subjects
0301 basic medicine ,Untranslated region ,Aging ,ABCE1 ,Medical Physiology ,Ribosome Recycling Factor ,translation ,Inbred C57BL ,Transgenic ,Mice ,0302 clinical medicine ,oxidative stress ,Aging brain ,3' Untranslated Regions ,Cellular Senescence ,Neurons ,MRNA cleavage ,Brain ,Translation (biology) ,Mitochondria ,Cell biology ,ribosome ,Neurological ,Female ,ribosome recycling ,3′ UTR ,brain ,mRNA ,1.1 Normal biological development and functioning ,Mice, Transgenic ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Underpinning research ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Gene ,Messenger RNA ,Three prime untranslated region ,Neurosciences ,Mice, Inbred C57BL ,Neostriatum ,Oxidative Stress ,030104 developmental biology ,Gene Expression Regulation ,NIH 3T3 Cells ,biology.protein ,ATP-Binding Cassette Transporters ,Biochemistry and Cell Biology ,Ribosomes ,030217 neurology & neurosurgery - Abstract
Particular brain regions and cell populations exhibit increased susceptibility to aging-related stresses. Here, we describe the age-specific and brain-region-specific accumulation of ribosome-associated 3′ UTR RNAs that lack the 5′ UTR and open reading frame. Our study reveals that this phenomenon impacts hundreds of genes in aged D1 spiny projection neurons of the mouse striatum and also occurs in the aging human brain. Isolated 3′ UTR accumulation is tightly correlated with mitochondrial gene expression and oxidative stress, with full-length mRNA expression that is reduced but not eliminated, and with production of short 3′ UTR-encoded peptides. Depletion of the oxidation-sensitive Fe-S cluster ribosome recycling factor ABCE1 induces the accumulation of 3′ UTRs, consistent with a model in which ribosome stalling and mRNA cleavage by No-Go decay yields isolated 3′ UTR RNAs protected by ribosomes. Isolated 3′ UTR accumulation is a hallmark of brain aging, likely reflecting regional differences in metabolism and oxidative stress. Particular brain regions and cell populations exhibit increased susceptibility to aging-related stresses. Sudmant et al. report that fragments of mRNAs accumulate in the aging brains of mice and humans. These species are associated with ribosomes and the production of small peptides and reflect regional differences in metabolism and oxidative stress., NIH (Grant HG002439)
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.